Next Article in Journal
The Role of Beta-Adrenergic Receptors in Depression and Resilience
Next Article in Special Issue
Signaling Switching from Hedgehog-GLI to MAPK Signaling Potentially Serves as a Compensatory Mechanism in Melanoma Cell Lines Resistant to GANT-61
Previous Article in Journal
Insulin Resistance and High Blood Pressure: Mechanistic Insight on the Role of the Kidney
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Modulation of Hedgehog Signaling for the Treatment of Basal Cell Carcinoma and the Development of Preclinical Models

Departments of Chemistry, Molecular Biosciences, Neurobiology, and Radiology Northwestern University, Evanston, IL 60208, USA
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(10), 2376; https://doi.org/10.3390/biomedicines10102376
Submission received: 30 August 2022 / Revised: 17 September 2022 / Accepted: 20 September 2022 / Published: 23 September 2022
(This article belongs to the Special Issue Targeting the Hedgehog Signaling Pathway in Cancer)

Abstract

:
Basal Cell Carcinoma (BCC) is the most commonly diagnosed cancer worldwide. While the survivability of BCC is high, many patients are excluded from clinically available treatments due to health risks or personal choice. Further, patients with advanced or metastatic disease have severely limited treatment options. The dysregulation of the Hedgehog (Hh) signaling cascade drives onset and progression of BCC. As such, the modulation of this pathway has driven advancements in BCC research. In this review, we focus firstly on inhibitors that target the Hh pathway as chemotherapeutics against BCC. Two therapies targeting Hh signaling have been made clinically available for BCC patients, but these treatments suffer from limited initial efficacy and a high rate of chemoresistant tumor recurrence. Herein, we describe more recent developments of chemical scaffolds that have been designed to hopefully improve upon the available therapeutics. We secondly discuss the history and recent efforts involving modulation of the Hh genome as a method of producing in vivo models of BCC for preclinical research. While there are many advancements left to be made towards improving patient outcomes with BCC, it is clear that targeting the Hh pathway will remain at the forefront of research efforts in designing more effective chemotherapeutics as well as relevant preclinical models.

Graphical Abstract

1. Introduction

Keratinocyte cancers, or nonmelanoma skin cancers (NMSCs), are the most commonly diagnosed cancers worldwide [1,2]. In the United States alone, one in every three to five Caucasian people are expected to develop an NMSC in their lifetime, with estimates as high as 4 million cases diagnosed each year [3,4,5]. Approximately 80% of all NMSCs are characterized as basal cell carcinomas (BCC), where uncontrolled growth of the basal cell population of the epidermis leads to tumorigenesis (Scheme 1) [6,7]. The overwhelming number of BCC diagnoses requires ample research and medical attention for the development of effective treatment and prevention strategies.
Gorlin Syndrome (GS) is a rare autosomal dominant disease comprising a small percentage of the BCC community and approximately 0.05% of the population [8]. In total, 90% of patients with GS experience the uncontrolled growth of multiple BCCs alongside various developmental abnormalities including those associated with holoprosencephaly and malignant medulloblastomas [8,9]. Sporadic BCC accounts for the predominant population of BCC patients. The primary cause of sporadic BCC is prolonged exposure to ultraviolet (UV) radiation from the sun [10,11]. The risk of developing BCC increases with light-skin pigmentation, age, and sunburn frequency during youth [12]. Other risk factors include family history of melanoma, blonde/red hair phenotype, and men are more susceptible to BCC than women [12,13].
A significant burden to the BCC patient community is the high rate of recurrence. BCC is clinically designated as low or high risk depending on the likelihood of recurrence [14]. However, BCC more commonly recurs in an entirely different location on the body. For primary tumor locations, the recurrence rate depends heavily on the method of treatment (discussed in depth in Section 2), with a range from 1 to 70% after 5 years [15,16]. Larger tumors also experience an increased likelihood of relapse [16]. More strikingly, the three-year risk of developing a second BCC lesion is estimated between 41–44% [15,17,18,19], and the likelihood increases with each additional lesion. Once diagnosed, approximately 50% of patients will battle BCC again.
BCC is classified in three primary identities: superficial (10–30%), nodular (60–80%), and morpheaform/infiltrative (<10%) [7,20]. Each differ in physical and histopathological behavior and exhibit differential relapse rates [21]. Superficial and nodular BCCs are less likely to recur, whereas infiltrative BCCs are more challenging to treat permanently [22]. Additionally, the different subtypes have variable occurrence rates on different skin areas. Nodular BCC is most commonly found on the face, while superficial BCC affects the torso and hands more frequently [23]. Infiltrative BCC is the most aggressive form and can often lead to the destruction of nearby healthy tissue [24]. Due to these differences in risk classification and behavior, selective care must be taken when deciding which treatment option to pursue for an individual BCC patient.
BCC’s overall survivability is very high, with estimates for mortality being less than 0.5% [25,26]. However, the exceptionally high number of BCC diagnoses means that even a low mortality rate produces significant BCC-related cancer deaths [5]. The American Cancer Society estimates this population at around 2000 NMSC-related deaths annually, primarily attributed to metastatic BCC complications.
With ever-increasing rate of BCC diagnoses, vast differences in subtypes, and extremely high rate of recurrence, it is imperative to focus BCC research efforts on preventing disease and improving disease outcomes. In this review, we outline the current state of research progress towards improving BCC treatment with a focus on the molecular drivers of BCC pathology. We first outline the treatment strategies employed in the clinic for BCC and highlight their advantages, disadvantages, and any known patient restrictions. Secondly, we elaborate on the Hedgehog (Hh) signaling cascade, the molecular driver of BCC. Thirdly, we provide an in-depth review of molecular chemotherapeutics that target the Hh pathway and how genetic modulation of Hh regulators has been used to develop in vivo BCC models. Finally, we provide a prospective on the state of the field and present opinions on future research priorities.

2. Predominant Treatment Options for BCC

Treatment strategies for BCC vary by subtype of the disease, size of the lesion, and patient age and preference. While the following list does not represent every option for BCC therapy, several predominant options are discussed in this section. Table 1 outlines the advantages and disadvantages of each, including any imperative patient restrictions.

2.1. Surgical Resection

Surgical intervention is by far the preferred treatment option for BCC owing to the highest rate of complete tumor clearance and lowest recurrence rates [15]. Two common forms of surgical intervention are wide local excision (WLE) and Mohs micrographic surgery (MMS). WLE relies on over-estimating the boundary of a tumor to completely remove it in a single surgical pass. While this can be effective for tumors with well-defined margins, many BCCs are more complex with unpredictable margins [27]. WLE can result in recurrence rates as high as 50% over 10 years if a tumor is not entirely excised [28].
MMS was first described in 1941 by Dr. Frederic Mohs and is used to treat many skin malignancies [29,30]. The surgery involves horizontally shaving a lesion in thin sections and evaluating each section by microscopy to detect cancerous tissue. Further excision is performed only where the tumor remains detected [31]. It promotes the identification of complete tumor margins while minimizing non-diseased tissue removal [32]. For BCC specifically, Mohs surgery is more effective than WLE in preventing tumor recurrence in both primary and recurrent lesions [30,33]. MMS is recommended for BCCs that exhibit more aggressive behavior and are situated at cosmetically sensitive locations to reduce disfiguration of the patient [34]. However, Mohs surgery requires highly skilled and trained surgeons on this complex technique and may be less available to patients in underdeveloped regions [28]. Additionally, some patients may refuse surgical intervention for personal reasons or are medically excluded as candidates. Of primary concern are elderly patients who may not properly recover from surgery and the potential cosmetic consequences of such invasive surgical procedures.

2.2. Radiation Therapy

BCC patients that cannot undergo or refuse surgical treatment for BCC require other therapeutic options. For example, lesions on the eyelids, nose, lips, and ears can be extremely challenging to completely excise surgically. Attempted excision may result in compromised anatomical function alongside undesired cosmetic outcomes [35]. Radiation therapy has emerged as one alternative in these situations, primarily due to its ability to treat both low- and higher-risk tumors [14]. Overall, the recurrence rate following radiation therapy alone is less than 10% over 5 years [36]. Radiotherapy is also helpful for cooperative treatment following surgery when margins are poorly defined or excision is incomplete [37].
Radiotherapy does, however, have limitations that are necessary to consider. Efficacy is dependent on tumor size and should be reserved for smaller tumors [28]. While older patients that are not surgical candidates can benefit significantly from radiotherapy, younger patients may experience an undesired cosmetic decline over time [36]. Any treatment with ionizing radiation increases the likelihood of developing other cancers, including melanoma. The destruction of tumor tissue cannot be guaranteed or determined without a more invasive follow-up. Most importantly, radiotherapy is rarely recommended for patients with GS [38]. Treatment with radiotherapy, even for non-skin cancer GS symptoms, may induce rampant BCC growth [39]. The complete destruction of tumor tissue cannot be guaranteed or determined without a more invasive follow-up.

2.3. Laser Therapies

Low-risk nodular and superficial BCCs may be non-invasively treated using laser-based techniques. Ablative therapy can be performed by carbon dioxide (CO2) or doped yttrium aluminum garnet (YAG) lasers [40]. Recurrence rates for this type of therapy are remarkably low at less than 3% [41]. Patients generally report favorable cosmetic outcomes compared to surgical resection [42], though it is unclear if this is due to inherent differences in tumor type. Mohs surgery is rarely used for smaller, lower-risk nodular/superficial BCCs, but laser treatments are. Scarring is logically expected to be less extreme for treatment of a smaller tumor.
Another laser-based technique for BCC treatment is photodynamic therapy (PDT). The mechanism of PDT is well characterized and, truthfully, most beneficial for treating skin cancers. A photosensitizer is delivered either topically or systemically to a tumor and irradiated with visible light that matches the excitation frequency of the sensitizer. The excited state transfers energy to water, producing cytotoxic singlet oxygen (1O2) and other reactive oxygen species (ROS) (Scheme 2) [28]. Due to the limited depth penetration of visible light, PDT has struggled to gain clinical approval to treat most cancers. However, PDT is an approved treatment for BCC in 18 countries [43,44]. In particular, superficial BCC has been extensively studied as a model cancer for evaluating PDT efficacy [44].
The safety profile of PDT is ideal for a tumor with such a high recurrence rate. The laser light is not harmful to the genetic composition of healthy tissue and is locally administered with high precision to the sensitizer-bearing tumor. PDT, however, carries some of the highest variability in treatment efficacy, with cure rates between 50–90% for primary tumors and as low as a 20% cure rate for recurrent tumors [16]. PDT can cause severe sensitivity to the sun post-treatment and may take several rounds to be maximally effective [45]. In contrast to radiotherapy, PDT is safe for patients with GS and effective in lesions with less than 2 mm depth (deeper tumors are not treatable by PDT due to limited tissue penetration of laser light) [46,47]. Additional research and optimization are required to improve the general efficacy of PDT against BCC.

2.4. Imiquimod and 5-Fluorouracil Topical Treatments

Imiquimod (Aldara; 3M Pharmaceuticals, Figure 1, left and 5-fluorouracil (Adrucil®, 5-FU, Figure 1, right are topical creams applied to BCC lesions for chemotherapeutic treatment. Imiquimod is approved by the Food and Drug Administration (FDA) for treating superficial BCCs less than 2 cm in diameter and is being evaluated for efficacy in nodular BCCs [48]. Therapeutic effect is achieved through an immune response and induction of apoptosis [49]. 5-FU is approved by the FDA for the treatment of superficial BCCs. The inhibition of nucleic acid synthesis is the primary mechanism of action. One study reported a 90% cure rate, with patients experiencing minimal side effects [50]. In both therapies, intense skin reactions are observed due to inflammation caused by imiquimod and lack of tumor specificity of 5-FU [28,51,52]. However, both therapies were found to be most effective when used in conjunction with other treatment options, suggesting that their utility might be optimized synergistically [51,53]. Topical treatment is ideal for skin cancers- localized delivery minimizes the harm to healthy tissues, specifically parts of the body completely unaffected by cancer. However, superficial BCC represents only 10–30% of the BCC patient population, severely limiting the number of patients who can access these treatments.

3. The Hedgehog Signaling Cascade in BCC

In the 1990s, genetic evaluation of BCCs of patients with GS revealed the most crucial discovery in BCC research history: BCC lesions are often linked to mutations in the patched1 (PTCH1) gene loci [54,55,56,57]. Since then, it has become commonly accepted that the Hedgehog (Hh) signaling cascade, to which PTCH1 proteins belong, is BCC’s primary oncogenic driver [58,59,60]. The Hh pathway is canonically activated (Figure 2) by the binding of Hh ligands to the transmembrane protein PTCH1, which releases smoothened (SMO) inhibition. Suppressor of fused (SUFU) is signaled to release glioma-associated oncogene (Gli) transcription factors where they are activated in the cytosol. Translocation into the nucleus activates the expression of target genes for cellular processes such as proliferation and migration [61,62]. Dysregulation of this pathway is associated with many cancers but is causative of BCC [58,59,60,63,64]. As such, it is a promising chemotherapeutic target for BCC.
Approximately 90% of sporadic BCCs arise from mutations of one PTCH1 allele, and 10% harbor mutations to downstream protein SMO [65]. Mutations in tumor suppressor p53 (p53) are also observed in BCC [66]. These mutations are consistent with genetic modifications commonly caused by UV exposure that ultimately leads to increased proliferation, maintained stemness, and tumorigenesis [23,65,67,68,69]. Additionally, activation of Hh signaling is often associated with the overexpression of programmed cell death ligand (PD-L1), promoting immunogenic escape and tumor cell proliferation [70,71].

4. Chemotherapies That Target Hedgehog Signaling

While treatment for lower-risk BCCs is mainly successful across the treatments described above, patients who suffer from high-risk infiltrative BCCs have fewer treatment options [72]. Infiltrative BCCs are broken further into locally advanced BCCs (laBCC) and metastatic BCCs (mBCC), and often, surgical resection is not an option for these patients. While mBCC only occurs in less than 0.5% of cases [72,73], it presents a unique treatment challenge. The majority of metastasis is observed in the lymph nodes, lungs, liver, and bone [72]. Before the development of systemic chemotherapies targeting Hh signaling, median survivability for mBCC patients was only 8 months after diagnosis [74].

4.1. Smoothened Inhibitors

The overwhelming majority of Hh-specific therapies target the transmembrane protein SMO. The first described SMO antagonist is Cyclopamine (Figure 3A), a natural product found in corn lily [75]. Pregnant ewes grazed on corn lily produced offspring with craniospinal defects, including cyclopia, that could not be explained [76,77]. During this time, a connection between mutations in the Hh pathway genome and the occurrence of holoprosencephaly (including cyclopia) in mammals was found [78,79]. After extensive research, inhibition of SMO by what is now commonly referred to as Cyclopamine was determined to cause birth defects in the ewe litters [80,81]. While studied extensively as a chemotherapeutic agent [82,83,84,85,86,87,88], Cyclopamine suffers from poor bioavailability due to a lack of solubility and stability [75]. However, the structural elucidation of Cyclopamine promoted the development of analogs with improved biocompatibility.
Two SMO inhibitors have received FDA approval for the treatment of advanced BCCs. Vismodegib (GDC-0449, Erivedge®, Figure 3B) is approved to treat recurrent, locally advanced, and metastatic BCCs in patients who are not candidates for surgery or radiation therapy [89]. Sonidegib (Odomzo, Novartis, Figure 3C) is approved for laBCC in patients who are not candidates for surgery or radiation therapy [90]. Both inhibitors have shown efficacy for some patients whose outcomes might have otherwise been poor, but they certainly are a far cry from the perfect answer to BCC treatment. Approximately 50% of patients treated with Vismodegib have no initial response, and of those that do, over 20% develop chemo-resistant tumor recurrence [91,92]. Many of the mutations that lead to chemoresistance were identified in the drug target, SMO, suggesting that mutations of SMO structure in primary tumors may explain the lack of response experienced by some patients [93,94,95]. Chemoresistant tumor recurrence is a significant issue considering that BCC already exhibits such high recurrence rates. Vismodegib is administered orally as this is the only way to ensure that metastases are effectively treated but means that all areas of the body are exposed to the drug. Secondary BCC lesions that could grow in new locations might also develop resistance to further Vismodegib treatment. Additionally, patients receiving these treatments often experience untoward side effects such as muscle cramps, loss of taste, weight loss, hair loss, and mental health decline that are not always amenable to continued therapy [96,97]. Muscle cramps are most common due to the activation of calcium flux upon inhibition of canonical Hh signaling. Other symptoms such as hair and taste loss stem from the systemic inhibition of Hh signaling required for the maintenance of hair follicles and taste buds. Approximately 20% of BCC patients enrolled in various trials with SMO inhibitors discontinue treatment due to these side effects [97].
Figure 3. Chemical structures of smoothened inhibitors (A) Cyclopamine. (B) Vismodegib. (C) Sonidegib. While distantly related to Cyclopamine, Vismodegib and Sonidegib contain structural similarities. All three inhibitors bind SMO in the same pocket. (D,E) New generation Vismodegib derivatives developed by Li et al. in 2019 [98]. (F) To date, the most potent SMO inhibitor reported by Hoch, et al.
Figure 3. Chemical structures of smoothened inhibitors (A) Cyclopamine. (B) Vismodegib. (C) Sonidegib. While distantly related to Cyclopamine, Vismodegib and Sonidegib contain structural similarities. All three inhibitors bind SMO in the same pocket. (D,E) New generation Vismodegib derivatives developed by Li et al. in 2019 [98]. (F) To date, the most potent SMO inhibitor reported by Hoch, et al.
Biomedicines 10 02376 g003
However, chemoresistant recurrence remains a serious concern. In one study, researchers attempted to treat Vismodegib resistant tumors with Sonidegib to test the hypothesis that a different SMO inhibitor might still be effective. The results of this study concluded that patients with Vismodegib resistance are likely for their disease to continue to progress if treated with Sonidegib, specifically [96]. A limitation of this study is that Vismodegib and Sonidegib have similar chemical structures and bind SMO in the same location [96]. It cannot be concluded that all SMO inhibitors would be ineffective, but only that Sonidegib was insufficient to overcome Vismodegib resistance.
Recent studies have expanded upon structural components of Vismodegib that have resulted in more effective therapeutics and attenuation of resistance [98,99]. One study identified two new molecules as potent SMO inhibitors that are loosely founded upon the structure of Vismodegib. These compounds labeled HH-13 (Figure 3D) and HH-20 (Figure 3E) displayed 10 and 30 nM IC50 values in cellular assays of Hh activity, respectively [98]. Most importantly, these compounds remained effective against SMO-D473H, a SMO mutant that Vismodegib is incapable of inhibiting. Vismodegib efficacy is diminished by almost 1000-fold between wild type SMO and SMO-D473H, whereas the effectiveness of HH-13 and HH-20 is only decreased by 1.1 and 1.4-fold, respectively. While this is an exciting advancement in the realm of development of SMO inhibitors, only one mutant version of SMO was evaluated. It is likely that HH-13 and HH-20 will not be effective against all SMO mutants.
To date, the most effective SMO inhibitor was first described by Hoch et al. in 2015 and inhibits SMO with approximately 10-fold improvement in potency over Vismodegib. MRT-92 (Figure 3F) is an acylguanidine derivative with structural differences from Vismodegib that promote binding to a different locale in the SMO structure. Whereas Vismodegib binds SMO in the extracellular domain, the MRT-92 scaffold was shown to bind the entire length of the SMO transmembrane domain. This provides a competitive advantage against common SMO mutations. MRT-92 remains potent against the SMO-D473H mutation due to retained binding affinity where Vismodegib binding affinity is lost entirely [100]. Additionally, MRT-92 successfully controlled the tumor growth of a murine xenograft melanoma model, suggesting applicability to BCC [101].
Another possible solution to improve SMO inhibition is the exploration of chemical structures that deviate from that of Vismodegib. In 2015, the Waldmann group identified that synthetic modifications to the natural product withaferin A (Figure 4A) produced potent inhibitors of SMO. Specifically, compound 21a (Figure 4B) exhibited a strong binding affinity for SMO and an IC50 around 2 µM [102]. However, the synthesis of these complexes is non-trivial and is stereoselective. Diastereomerization of 21a to 21a’ (Figure 4C) reduced potency by almost 5-fold [102].
In 2021, the Passarella group proposed simplifying the structure to contain cyclic carbamates with the ultimate goal of synthesizing and evaluating compound 1 (Figure 4D) [103]. However, stereoisomers of this compound ultimately proved inactive against Hh signaling. Two pathway intermediates, 13b (Figure 4E) and 14b (Figure 4F), successfully inhibited SMO with racemic IC50 values of 7.4 µM and 13.0 µM, respectively. Enantioselective synthesis revealed that (+)-13b and (−)-14b were the more potent inhibitors with IC50 values around 6 µM compared to their enantiomers at 11–16 µM [103]. While it is surprising that deprotection of 14b to yield compound 1 eliminates activity, the tert-butyl(chloro)diphenylsilane (TBDPS) protecting group is highly lipophilic and might significantly impact protein interaction. While the potency of these complexes does not compare to clinically available Vismodegib and Sonidegib, further study is necessary to evaluate inhibition in BCC specifically and the ability to evade resistance.

4.2. Gli Inhibitors

While newer generations of SMO inhibitors with structurally diverse scaffolds are promising, it is unclear if they will successfully evade the complications of the already approved compounds. As mentioned above, mutations of SMO itself render continued treatment with SMO inhibitors ineffective if the mutation abolishes drug binding. Additionally, cellular switches have been identified to bypass SMO activity in some recurrent tumor pathology [104]. Therefore, options targeting other Hh signaling regulators would be beneficial for patients who do not initially respond to treatment with SMO inhibitors or develop resistance. In one study addressing Vismodegib resistance, mutations of SMO proteins were the primary focus. Two smoothened variants known to be insensitive to Vismodegib were expressed in SMO knockout mouse embryonic fibroblasts. Upon treatment with both direct and indirect Gli transcription factor inhibitors, Hh activity was indeed reduced regardless of the identity of the SMO mutation [94]. In another study, activation of serum response factor and the transcriptional cofactor megakaryoblastic leukemia 1/2 (MKL1/2) were found to have a novel, non-canonical interaction with Gli1 that amplified Hh transcription independently of SMO. Excellent in vivo anticancer activity was achieved through MKL1/2 inhibition in Vismodegib resistant tumors with this characteristic [105]. Interestingly, both the canonical and non-conical resistance mechanisms ultimately influence the activity of Gli1 transcription factors in a way that is druggable [106]. As such, Gli is a valuable target for chemotherapeutic intervention in BCC.
To date, no therapies targeting the Gli family of zinc finger transcription factors (ZnFtfs) have received clinical approval. This is primarily attributed to the fact that transcription factors, in general, are notoriously challenging to target specifically with traditional small molecules due to a lack of well-defined binding pockets [107,108,109]. However, a few small molecule inhibitors for Gli proteins have been developed and studied against Hh signaling. Figure 5 depicts the inhibitors discussed in this section.
One of the first small molecules found to inhibit Hh signaling through Gli downregulation is arsenic trioxide (ATO, Figure 5A) [110]. Importantly, ATO is effective in the treatment of tumors that have developed SMO resistance around a dose of 500 nM [111]. However, ATO is not specific to Gli transcription factors and is known to bind numerous intracellular targets [112]. In fact, it is FDA approved (Trisenox, Cell Therapeutics) for the treatment of acute myeloid leukemia due to its ability to potently inhibit promyelogenous leukemia-retinoic acid receptor fusion protein [113,114]. Ideally, a Gli inhibitor would be both potent and specific to reduce unwanted off-target complications.
The most prominent small molecule Gli inhibitor is GANT-61, a derivative of the Gli antagonist (GANT) family of compounds. It was discovered in 2007 and has since been used to study Hh inhibition in a variety of cancers [115,116,117,118,119,120,121,122]. The general inhibitory concentration at which 50% of Hh signaling is reduced (IC50) is on the order of 5–10 µM for GANT-61 [123,124]. GANT-61 is understood to undergo a prodrug mechanism where hydrolysis of the intact molecule produces an inactive side product and the active inhibitor (Figure 5B) [125]. Computational analysis further suggests a direct binding mechanism of GANT-61 to Gli transcription factors that inhibits DNA binding and therefore transcription, but this has yet to be confirmed experimentally [125,126,127].
Ultimately, GANT-61 is limited by poor solubility and bioavailability [128]. More recently, the natural product Glabrescione B (GlaB, Figure 5C) was reported as the first confirmed small molecule to directly bind Gli and prevent the Gli/DNA binding interaction. GlaB inhibited BCC growth in vitro and in vivo at the equivalent of low µM doses [129]. When compared directly to GANT-61 in this work, GlaB was found to have no significant improvement in potency or inhibitory effect. However, GANT-61 efficacy was only directly compared with in vitro experiments, not in vivo. It is possible that GlaB exhibits higher bioavailability and would be more effective in vivo. However, a study of GlaB against Hh activity in medulloblastoma showed that micelle encapsulation improved solubility and potency, revealing that GlaB efficacy similarly suffers from low bioavailability [130].
The discovery of Gli inhibition by natural product GlaB suggests potential core chemical structures that could by synthetically modified to improve solubility and binding to Gli transcription factors. Specifically, modifications to the isoflavone core (Figure 5C,D) have been made to study the effect of structure on efficacy. Chemical modifications at the meta and para positions of the third ring generate compounds that influence inhibitory potential. Compounds 5 and 12 (Figure 5D) inhibit Gli with IC50 values of in the 2–10 µM range, similar to GlaB [131]. However, compound 17 did not show inhibitory potential under 30 µM, suggesting that very small structural changes make large differences in protein binding.
Further chemical modification of this scaffold has revealed that bulky substituents at the meta position produce isoflavones that target Gli, but bulky substituents at the para position generate compounds that target SMO [131,132]. Combining these principles yielded compound 22 (Figure 5E) which targets both SMO and Gli. This agent successfully inhibited tumor growth in a model of medulloblastoma. However, the ability to target two proteins means that selectivity for this compound is questionable. It is unknown what other molecular targets it may bind, producing unwanted off-target effects.
In 2020, a new structural scaffold was reported to inhibit Gli transcription factors. Bicyclic imidazolium compounds were first discovered to inhibit Gli in a high throughout drug screen and then were evaluated across a range of structure/activity relationship studies [133]. Of 12 synthesized molecules, compound 10 (Figure 5F) was found to have the highest anti-Gli activity with an IC50 between 100 nM and 5 µM, depending on the assay. Structural considerations determined that the terminal phenyl group added significant potency to the molecules, whereas adding heteroatoms into the bi/tricyclic ring system essentially eliminated anti-Gli activity [133]. It is worthy to note that these complexes are highly aromatic and hydrophobic, much like GANT-61 and GlaB. It is likely that solubility will continue to be an issue for these compounds, though this was not discussed by the authors. Additionally, these complexes significantly interfered with mitochondrial health and function, something that must be considered within the context of selectivity and unwanted side effects in non-diseased tissue.
The discovery of cisplatin as an anticancer agent birthed an entirely new field of medicinal inorganic chemistry [134,135]. Inorganic compounds can be desirous as therapeutics for their ability to access inhibitory mechanisms unknown to organic compounds. In recent studies, the zinc ions that structurally support the alpha-helical structure of Gli transcription factors were targeted by a cobalt-Schiff base inorganic complex (Figure 6A) [136,137]. Cobalt-Schiff base complexes have been shown to displace the zinc ions from Cis2His2 coordination packets via preferential histidine binding of cobalt [138]. The alpha-helical structure of Gli is then depleted in the DNA binding domain. As a result, the Gli/DNA interaction is inhibited, and target genes would not be transcribed [137].
To achieve specificity, the consensus DNA sequence that only one transcription factor will recognize is tethered to the active cobalt-Schiff base inhibitor. This brings the cobalt complex into close enough proximity to the protein to irreversibly displace zinc ions. This strategy has been previously employed in Drosophila and Xenopus model organisms of homologous Hh pathways [139,140,141]. CoGli (Figure 6B) was developed by Dukes et al. to inhibit the Gli family of transcription factors in cellular assays of BCC. In this study, GANT-61 served as a positive control. GANT-61 inhibited Hh-driven migration of the ASZ murine BCC cell line by approximately 50% at a 5 µM dose. Strikingly, the targeted cobalt-DNA complex delivered by a cationic vehicle inhibited Hh-driven migration by 50% at only 300 nM [136]. This represents a promising new direction in the field of Gli inhibition as the first inhibitor to exhibit nM efficacy with a high degree of selectivity.
While GANT-61 has seemingly exhausted its potential for clinical translation, GlaB derivatives and imidazolium compounds show potential for further development as potent organic Gli inhibitors. Cobalt-DNA based complexes show additional promise with unique inhibition mechanisms and improved target selectivity. However, few of these compounds have been thoroughly evaluated for Hh inhibition in BCC, specifically. It is challenging to conclude their applicability to BCC treatment but highlights the need for further study and development for an effective Gli inhibitor to achieve clinical approval.

5. Preclinical Models for Hedgehog and BCC Research

A significant challenge for the BCC research community is the overall paucity of preclinical models for evaluating therapeutics. This is even more problematic when considering the unavailability of human-derived models. While animal models are broadly used as research tools in therapeutic development, fundamental differences between different species directly impact the translation of therapeutics. Here, we outline the current and recent therapeutic evaluation developments in both Hh activity cell lines and BCC-specific model systems.

5.1. Hedgehog Activation in Cellular Assays

The first benchmark for evaluating a new Hh inhibitor is often a cellular assay of Hh activity. One example is a luciferase reporter assay performed in a derivative of the NIH-3T3 mouse fibroblast cell line that contains modified Gli binding domains driving expression of firefly luciferase [142]. When Hh signaling is exogenously activated, luciferase is stably expressed and can be measurably down-regulated by concurrent treatment with Hh inhibitors [143,144,145]. The pathway can be activated at PTCH1, SMO, or by transfecting a plasmid encoding for Gli transcription factors to model Gli accumulation.
The C3H/10T1/2 cell line is a pluripotent mouse embryonic fibroblast routinely used in Hh research [146]. The cells do not exhibit innate Hh activity, but exogenous activation promotes differentiation into osteoblasts and induces alkaline phosphatase (ALP) protein expression [147,148,149]. Concurrent treatment with Hh inhibitors results in measurable prevention of ALP production. This cellular system has dramatically increased the understanding of basic Hh mechanics and the general efficacy of inhibition strategies [150]. However, both NIH-3T3 and C3H/10T1/2 cellular assays are not representative of Hh dysregulation in a tumor environment and are also not derived from skin cells. While suitable for initial evaluation of Hh inhibitory potential, more specific models are necessary to evaluate applicability to BCC.

5.2. Murine BCC Models

Rodents (mice and rats) are choice mammals for most early-stage preclinical investigations of cancer treatments. Developing a rodent model that most closely mimics human disease is essential for successful translation into the clinic. As such, it is desirous for an animal to grow tumors spontaneously. One of the first mouse models to spontaneously developed BCC was generated by overexpressing sonic hedgehog (SHH) proteins that initiate Hh signaling [151]. SHH is a paracrine signal, however, and activation was not isolated to skin cells malformations across the animal were observed.
Additionally, the animals had to be examined either in the embryonic or neonatal states due to uncontrollable perinatal lethality. While embryos did develop large BCC-like lesions that mimicked patient BCC phenotype and pathology, they most closely resembled uncontrollable GS BCC growth. Animals allowed to grow long enough to die in utero had large sections of skin destroyed from advanced disease [151]. Ultimately, the untimely death of the animals in this study prevents the development of a breedable line that could be used to investigate Hh inhibitors. However, it provides evidence for the ability to develop murine BCC models through manipulation of the Hh genome.
Other studies attempted a similar generation of spontaneous BCC models through transgenic Hh activation. One report induced expression of a constituently active mutant of SMO under a keratin 5 promoter that confined expression to the skin. BCCs developed in embryos that mimicked patient phenotype and pathology [152]. The authors do not comment on perinatal lethality, but further research determined that animals that do survive cannot reproduce to generate a breedable line [153]. However, localized expression in the skin avoided the craniospinal defects seen in SHH overexpression, reducing discomfort and suffering of surviving animals.
PTCH1 alleles are the most common source of mutations leading to sporadic BCCs in humans. As such, knockdown of Ptch1 has been attempted for the development of spontaneous BCCs in mice. An extensive review of Ptch1 knockout mice has been previously published [154]. Here, we focus on the broad story of development.
Early efforts towards this aim proved fruitless. Animals were viable but developed medulloblastomas [155] and rhabdomyosarcomas [156] and even other symptoms of GS but did not produce apparent BCCs. Further studies revealed that Ptch1 heterozygous mice at 9 months had small proliferations of BCC-like cells that could only be detected microscopically [153]. To encourage tumorigenesis, mice were subjected to UV or X-ray irradiation. After UV irradiation, Ptch1 knockout mice had a 20% incidence of developing lesions that mimicked the phenotype of human BCCs. X-ray irradiation produced trichoblastomas primarily.
The generation of BCCs from Ptch1 knockout mouse models resulted in an equally important development: BCC’s first immortalized cell lines. Three cell lines were isolated and immortalized from three different mouse models. The most commonly studied ASZ001, or ASZ, the cell line was immortalized from a BCC lesion resulting from UV irradiation three times weekly for 10 months [153]. These cells retain knockdown of Ptch1 in culture and are verified to be sensitive to Hh inhibition.
When irradiated, tumors on Ptch1 knockdown mice develop in a controlled manner where the UV light is applied. This significantly reduces the number of lesions from a truly spontaneous model and allows for a more controlled experimental design. Notably, some mice developed tumors in as little as four months of UV irradiation [153]. After histopathological validation of tumor type, these lesions could be treated with Hh inhibitors on a semi-reasonable time scale. However, the model produces a significant time burden from breeding to birth to tumor development. The answer to this rime delay for many cancers is patient-derived xenograft (PDX) models. Cells from human cancer patients are injected into an immunocompromised animal and develop into a tumor [157]. The time scale for this is exponentially faster and does not require the use of heavily genetically modified animals. Additionally, cancer is now fundamentally of human identity.
Unfortunately, developing PDX models of BCC has largely failed. Tumors often do not implant or are met with slow growth rates [158,159]. The use of more severally immunocompromised mice improves implantation [160,161], but little is known about the retention of cellular identity and behavior to the original tumor [162]. However, one study successfully allografted murine BCC cells into an immunocompromised mouse model with the assistance of Matrigel. Allografted tumors retained the phenotype and pathology of their parent tumors and were responsive to inhibition. Most importantly, allografts produced visible tumors within only 3 weeks of implantation [162]. This technique is unique and could be applied to the grafting of patient samples for a more human-based model of BCC.
As mentioned previously, there is an inherent distant relationship between the tissues of rodents and humans. Therefore, without available PDX models, a species more closely related to human identity is a valuable research tool. In 2017, a group investigated the ability to generate a BCC model in a non-human primate, the Chinese tree shrew [163]. Chinese tree shrews are small and have been used to study many human diseases [164]. Their skin is anatomically similar to human skin, creating a unique opportunity for more accurate BCC model development [165]. Development of the tree shrew model of BCC was accomplished via lentiviral transfection of SmoA1, a constitutively activated form of SMO. Injections were performed in both dorsal and tail skin, resulting in the development of BCC lesions that mimic human BCC. While this model has yet to be used to investigate BCC inhibition, it provides an interesting preclinical link between murine and human species.

6. Perspectives

While many BCC cases are readily cured via surgical methods, both ineligibility and personal choice may leave many BCC patients without effective treatment options. As this cancer affects millions of people worldwide, alternative solutions are desirable. Herein, we have described scientific advancements in the development of targeted therapies for SMO and Gli proteins involved in the Hedgehog signaling cascade. Hh inhibitors are desirous due to the causative relationship between dysregulated signaling and BCC oncogenesis and progression. However, very few inhibitors have successfully translated to the clinic.
The two Hh targeted therapies that have reached clinical approval target SMO, an upstream Hh regulator prone to chemo-resistant mutation. Additionally, cellular switches and non-canonical Gli activation often render long-term SMO treatment ineffective. Gli has been identified as an alternative target but is challenging to specifically inhibit as few chemical structures selectively interact with Gli. While developing new chemical structures recognized to bind Gli and novel inorganic inhibition strategies are encouraging, many aspects of drug bioavailability must be optimized before these agents can be translated to the clinic.
Upon considering the available in vitro and in vivo models of BCC, we also identify this area of the research field to be lacking. The growth of spontaneous BCC tumors is very slow in successful transgenic mouse models, significantly hindering the time in which a research study can be performed. Additionally, many of these systems require the exposure of the animals to UV irradiation that mimics a moderate sunburn multiple times a week for several months. For these experiments to be justified and fruitful, we identify the need for an in vitro assay that is more complex than simple 2-dimensional cellular assays and mimics the characteristics of BCC in tissue. For many cancers, this can be accomplished through the development of 3D spheroid cultures. For skin cancer specifically, researchers have succeeded in developing 3D epidermal mimics that can be assembled to mirror a skin cancer of interest [166]. We propose the development of a similar model of BCC to be of high utility to the research field at large.
Finally, the described preclinical models of BCC have only been used to evaluate the treatment of established disease. They have not been utilized to study how BCC latent potential ultimately leads to lesions. This should be a significant research endeavor considering BCC’s rate of primary and secondary tumor recurrence. Understanding these mechanisms in skin cells might elucidate new ways BCC can be prevented. All BCC patients should be considered at high risk for developing multiple BCCs, and effective prevention strategies would significantly improve the lives of BCC patients.

Author Contributions

This review article was written by M.W.D. and T.J.M. assisted with editing and final formatting. All authors have read and agreed to the published version of the manuscript.

Funding

M.W.D. and T.M. acknowledge NIH grants 1F31CA236175 and R01NS115571.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lai, V.; Cranwell, W.; Sinclair, R. Epidemiology skin cancer in the mature patient. Clin. Dermatol. 2018, 36, 167–176. [Google Scholar] [CrossRef] [PubMed]
  2. Albert, M.R.; Weinstock, M.A. Keratinocyte Carcinoma. CA Cancer J. Clin. 2003, 53, 292–302. [Google Scholar] [CrossRef]
  3. Stern, R.S. Prevalence of a history of skin cancer in 2007: Results of an incidence-based model. Arch. Dermatol. 2010, 146, 279–282. [Google Scholar] [CrossRef] [PubMed]
  4. Rogers, H.W.; Weinstock, M.A.; Feldman, S.R.; Coldiron, B.M. Incidence Estimate of Nonmelanoma Skin Cancer (Keratinocyte Carcinomas) in the U.S. Population, 2012. JAMA Dermatol. 2015, 151, 1081–1086. [Google Scholar] [CrossRef] [PubMed]
  5. Wong, C.S.M.; Strange, R.C.; Lear, J.T. Basal cell carcinoma. BMJ 2003, 327, 794–798. [Google Scholar] [CrossRef]
  6. Delishaj, D.; Rembielak, A.; Manfredi, B.; Ursino, S.; Pasqualetti, F.; Laliscia, C.; Orlandi, F.; Morganti, R.; Fabrini, M.G.; Paiar, F. Non-melanoma skin cancer treated with high-dose-rate brachytherapy: A review of literature. J. Contemp. Brachyther. 2016, 8, 533–540. [Google Scholar] [CrossRef] [PubMed]
  7. Cojocaru, A.; Marinescu, E.-A.; Nica, O.; Ilinoiu, E.; Negrila, A.; Ciurea, M.-E. Basal Cell Carcinoma and its Impact on Different Anatomical Regions. Curr. Health Sci. J. 2021, 47, 75–83. [Google Scholar] [CrossRef]
  8. Jones, E.A.; Sajid, M.I.; Shenton, A.; Evans, D.G. Basal Cell Carcinomas in Gorlin Syndrome: A Review of 202 Patients. J. Ski. Cancer 2011, 2011, 217378. [Google Scholar] [CrossRef]
  9. Kiwilsza, M.; Sporniak-Tutak, K. Gorlin-Goltz syndrome—A medical condition requiring a multidisciplinary approach. Med. Sci. Monit. 2012, 18, RA145–RA153. [Google Scholar] [CrossRef]
  10. Hoban, P.R.; Ramachandran, S.; Strange, R.C. Environment, phenotype and genetics: Risk factors associated with BCC of the skin. Expert Rev. Anticancer Ther. 2002, 2, 570–579. [Google Scholar] [CrossRef]
  11. Situm, M.; Buljan, M.; Bulat, V.; Lugović Mihić, L.; Bolanca, Z.; Simić, D. The role of UV radiation in the development of basal cell carcinoma. Coll. Antropol. 2008, 32 (Suppl. S2), 167–170. [Google Scholar]
  12. Wu, S.; Han, J.; Li, W.-Q.; Li, T.; Qureshi, A.A. Basal-cell carcinoma incidence and associated risk factors in U.S. women and men. Am. J. Epidemiol. 2013, 178, 890–897. [Google Scholar] [CrossRef]
  13. Berlin, N.L.; Cartmel, B.; Leffell, D.J.; Bale, A.E.; Mayne, S.T.; Ferrucci, L.M. Family history of skin cancer is associated with early-onset basal cell carcinoma independent of MC1R genotype. Cancer Epidemiol. 2015, 39, 1078–1083. [Google Scholar] [CrossRef]
  14. Puig, S.; Berrocal, A. Management of high-risk and advanced basal cell carcinoma. Clin. Transl. Oncol. 2015, 17, 497–503. [Google Scholar] [CrossRef]
  15. Chung, S. Basal cell carcinoma. Arch. Plast. Surg. 2012, 39, 166–170. [Google Scholar] [CrossRef]
  16. Bøgelund, F.S.; Philipsen, P.A.; Gniadecki, R. Factors affecting the recurrence rate of basal cell carcinoma. Acta Derm. Venereol. 2007, 87, 330–334. [Google Scholar] [CrossRef]
  17. Marcil, I.; Stern, R.S. Risk of developing a subsequent nonmelanoma skin cancer in patients with a history of nonmelanoma skin cancer: A critical review of the literature and meta-analysis. Arch. Dermatol. 2000, 136, 1524–1530. [Google Scholar] [CrossRef]
  18. Levi, F.; Randimbison, L.; Maspoli, M.; Te, V.C.; La Vecchia, C. High incidence of second basal cell skin cancers. Int. J. Cancer 2006, 119, 1505–1507. [Google Scholar] [CrossRef]
  19. Bartos, V. Development of Multiple-Lesion Basal Cell Carcinoma of the Skin: A Comprehensive Review. Sisli Etfal Hast. Tip Bülteni 2019, 53, 323–328. [Google Scholar] [CrossRef]
  20. Villani, R.; Murigneux, V.; Alexis, J.; Sim, S.-L.; Wagels, M.; Saunders, N.; Soyer, H.P.; Parmentier, L.; Nikolaev, S.; Fink, J.L.; et al. Subtype-Specific Analyses Reveal Infiltrative Basal Cell Carcinomas Are Highly Interactive with their Environment. J. Investig. Dermatol. 2021, 141, 2380–2390. [Google Scholar] [CrossRef]
  21. Cameron, M.C.; Lee, E.; Hibler, B.P.; Barker, C.A.; Mori, S.; Cordova, M.; Nehal, K.S.; Rossi, A.M. Basal cell carcinoma: Epidemiology; pathophysiology; clinical and histological subtypes; and disease associations. J. Am. Acad. Dermatol. 2019, 80, 303–317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Armstrong, L.T.D.; Magnusson, M.R.; Guppy, M.P.B. Risk factors for recurrence of facial basal cell carcinoma after surgical excision: A follow-up analysis. J. Plast. Reconstr. Aesthetic Surg. 2017, 70, 1738–1745. [Google Scholar] [CrossRef] [PubMed]
  23. Fania, L.; Didona, D.; Morese, R.; Campana, I.; Coco, V.; Di Pietro, F.R.; Ricci, F.; Pallotta, S.; Candi, E.; Abeni, D.; et al. Basal Cell Carcinoma: From Pathophysiology to Novel Therapeutic Approaches. Biomedicines 2020, 8, 449. [Google Scholar] [CrossRef] [PubMed]
  24. Hendrix, J.D., Jr.; Parlette, H.L. Duplicitous growth of infiltrative basal cell carcinoma: Analysis of clinically undetected tumor extent in a paired case-control study. Dermatol. Surg. 1996, 22, 535–539. [Google Scholar] [CrossRef]
  25. Wehner, M.R.; Cidre Serrano, W.; Nosrati, A.; Schoen, P.M.; Chren, M.-M.; Boscardin, J.; Linos, E. All-cause mortality in patients with basal and squamous cell carcinoma: A systematic review and meta-analysis. J. Am. Acad. Dermatol. 2018, 78, 663–672.e663. [Google Scholar] [CrossRef]
  26. Kim, D.P.; Kus, K.J.B.; Ruiz, E. Basal Cell Carcinoma Review. Hematol. Oncol. Clin. N. Am. 2019, 33, 13–24. [Google Scholar] [CrossRef]
  27. Quazi, S.J.; Aslam, N.; Saleem, H.; Rahman, J.; Khan, S. Surgical Margin of Excision in Basal Cell Carcinoma: A Systematic Review of Literature. Cureus 2020, 12, e9211. [Google Scholar] [CrossRef]
  28. Ceilley, R.I.; Del Rosso, J.Q. Current modalities and new advances in the treatment of basal cell carcinoma. Int. J. Dermatol. 2006, 45, 489–498. [Google Scholar] [CrossRef]
  29. Mohs, F.E. Chemosurgery: A microscopically controlled method of cancer excision. Arch. Surg. 1941, 42, 279–295. [Google Scholar] [CrossRef]
  30. Tolkachjov, S.N.; Brodland, D.G.; Coldiron, B.M.; Fazio, M.J.; Hruza, G.J.; Roenigk, R.K.; Rogers, H.W.; Zitelli, J.A.; Winchester, D.S.; Harmon, C.B. Understanding Mohs Micrographic Surgery: A Review and Practical Guide for the Nondermatologist. Mayo Clin. Proc. 2017, 92, 1261–1271. [Google Scholar] [CrossRef]
  31. Smeets, N.W.J.; Kuijpers, D.I.M.; Nelemans, P.; Ostertag, J.U.; Verhaegh, M.E.J.M.; Krekels, G.A.M.; Neumann, H.A.M. Mohs’ micrographic surgery for treatment of basal cell carcinoma of the face—Results of a retrospective study and review of the literature. Br. J. Dermatol. 2004, 151, 141–147. [Google Scholar] [CrossRef]
  32. Bittner, G.C.; Cerci, F.B.; Kubo, E.M.; Tolkachjov, S.N. Mohs micrographic surgery: A review of indications, technique, outcomes, and considerations. Bras. Dermatol. 2021, 96, 263–277. [Google Scholar] [CrossRef]
  33. Rowe, D.E.; Carroll, R.J.; Day, C.L., Jr. Mohs Surgery Is the Treatment of Choice for Recurrent (Previously Treated) Basal Cell Carcinoma. J. Dermatol. Surg. Oncol. 1989, 15, 424–431. [Google Scholar] [CrossRef]
  34. Lawrence, C.M. Mohs surgery of basal cell carcinoma—A critical review. Br. J. Plast. Surg. 1993, 46, 599–606. [Google Scholar] [CrossRef]
  35. Likhacheva, A.; Awan, M.; Barker, C.A.; Bhatnagar, A.; Bradfield, L.; Brady, M.S.; Buzurovic, I.; Geiger, J.L.; Parvathaneni, U.; Zaky, S.; et al. Definitive and Postoperative Radiation Therapy for Basal and Squamous Cell Cancers of the Skin: Executive Summary of an American Society for Radiation Oncology Clinical Practice Guideline. Pract. Radiat. Oncol. 2020, 10, 8–20. [Google Scholar] [CrossRef]
  36. Silverman, M.K.; Kopf, A.W.; Gladstein, A.H.; Bart, R.S.; Grin, C.M.; Levenstein, M.J. Recurrence rates of treated basal cell carcinomas. Part 4: X-ray therapy. J. Dermatol. Surg. Oncol. 1992, 18, 549–554. [Google Scholar] [CrossRef]
  37. Baheti, A.D.; Tirumani, S.H.; Giardino, A.; Rosenthal, M.H.; Tirumani, H.; Krajewski, K.; Ramaiya, N.H. Basal Cell Carcinoma: A Comprehensive Review for the Radiologist. Am. J. Roentgenol. 2015, 204, W132–W140. [Google Scholar] [CrossRef]
  38. Baker, S.; Joseph, K.; Tai, P. Radiotherapy in Gorlin Syndrome: Can It Be Safe and Effective in Adult Patients? J. Cutan. Med. Surg. 2016, 20, 159–162. [Google Scholar] [CrossRef]
  39. Lo Muzio, L. Nevoid basal cell carcinoma syndrome (Gorlin syndrome). Orphanet J. Rare Dis. 2008, 3, 32. [Google Scholar] [CrossRef]
  40. Salavastru, C.; Tiplica, G.S.; Fritz, K. Lasertherapie des Basalzellkarzinoms. Der Hautarzt 2018, 69, 10–16. [Google Scholar] [CrossRef]
  41. Moskalik, K.; Kozlov, A.; Demin, E.; Boiko, E. The efficacy of facial skin cancer treatment with high-energy pulsed neodymium and Nd:YAG lasers. Photomed. Laser Surg. 2009, 27, 345–349. [Google Scholar] [CrossRef] [PubMed]
  42. Ahluwalia, J.; Avram, M.M.; Ortiz, A.E. The Evolving Story of Laser Therapeutics for Basal Cell Carcinoma. Dermatol. Surg. 2020, 46, 1045–1053. [Google Scholar] [CrossRef] [PubMed]
  43. Wong, T.H.; Morton, C.A.; Collier, N.; Haylett, A.; Ibbotson, S.; McKenna, K.E.; Mallipeddi, R.; Moseley, H.; Seukeran, D.C.; Rhodes, L.E.; et al. British Association of Dermatologists and British Photodermatology Group guidelines for topical photodynamic therapy 2018. Br. J. Dermatol. 2019, 180, 730–739. [Google Scholar] [CrossRef] [PubMed]
  44. Collier, N.J.; Rhodes, L.E. Photodynamic Therapy for Basal Cell Carcinoma: The Clinical Context for Future Research Priorities. Molecules 2020, 25, 5398. [Google Scholar] [CrossRef]
  45. Szeimies, R.M.; Ibbotson, S.; Murrell, D.F.; Rubel, D.; Frambach, Y.; De Berker, D.; Dummer, R.; Kerrouche, N.; Villemagne, H.; on behalf of the Excilight Study Group. A clinical study comparing methyl aminolevulinate photodynamic therapy and surgery in small superficial basal cell carcinoma (8–20 mm), with a 12-month follow-up. J. Eur. Acad. Dermatol. Venereol. 2008, 22, 1302–1311. [Google Scholar] [CrossRef] [PubMed]
  46. Basset-Seguin, N.; Bissonnette, R.; Girard, C.; Haedersdal, M.; Lear, J.T.; Paul, C.; Piaserico, S. Consensus recommendations for the treatment of basal cell carcinomas in Gorlin syndrome with topical methylaminolaevulinate-photodynamic therapy. J. Eur. Acad. Dermatol. Venereol. 2014, 28, 626–632. [Google Scholar] [CrossRef]
  47. Marous, M.; Mueller, K.; Tausk, F. Multiple basal cell carcinomas in Gorlin Syndrome treated with pulsed dye laser. J. Cosmet. Laser Ther. 2020, 22, 230–231. [Google Scholar] [CrossRef]
  48. Shumack, S.; Robinson, J.; Kossard, S.; Golitz, L.; Greenway, H.; Schroeter, A.; Andres, K.; Amies, M.; Owens, M. Efficacy of Topical 5% Imiquimod Cream for the Treatment of Nodular Basal Cell Carcinoma: Comparison of Dosing Regimens. Arch. Dermatol. 2002, 138, 1165–1171. [Google Scholar] [CrossRef]
  49. Dummer, R.; Urosevic, M.; Kempf, W.; Hoek, K.; Hafner, J.; Burg, G. Imiquimod in basal cell carcinoma: How does it work? Br. J. Dermatol. 2003, 149 (Suppl. S66), 57–58. [Google Scholar] [CrossRef]
  50. Gross, K.; Kircik, L.; Kricorian, G. 5% 5-Fluorouracil Cream for the Treatment of Small Superficial Basal Cell Carcinoma: Efficacy, Tolerability, Cosmetic Outcome, and Patient Satisfaction. Dermatol. Surg. 2007, 33, 433–440. [Google Scholar] [CrossRef]
  51. Epstein, E. Fluorouracil paste treatment of thin basal cell carcinomas. Arch. Dermatol. 1985, 121, 207–213. [Google Scholar] [CrossRef]
  52. Marks, R.; Gebauer, K.; Shumack, S.; Amies, M.; Bryden, J.; Fox, T.L.; Owens, M.L. Imiquimod 5% cream in the treatment of superficial basal cell carcinoma: Results of a multicenter 6-week dose-response trial. J. Am. Acad. Dermatol. 2001, 44, 807–813. [Google Scholar] [CrossRef]
  53. Torres, A.; Niemeyer, A.; Berkes, B. Treatment of basal cell carcinoma using imiquimod 5% cream as an adjuvant therapy to Mohs micrographic surgery. J. Eur. Acad. Dermatol. Venereol. 2004, 30, 1462–1469. [Google Scholar]
  54. Gailani, M.R.; Bale, S.J.; Leffell, D.J.; DiGiovanna, J.J.; Peck, G.L.; Poliak, S.; Drum, M.A.; Pastakia, B.; McBride, O.W.; Kase, R.; et al. Developmental defects in Gorlin syndrome related to a putative tumor suppressor gene on chromosome 9. Cell 1992, 69, 111–117. [Google Scholar] [CrossRef]
  55. 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]
  56. 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]
  57. Klein, R.D.; Dykas, D.J.; Bale, A.E. Clinical testing for the nevoid basal cell carcinoma syndrome in a DNA diagnostic laboratory. Genet. Med. 2005, 7, 611–619. [Google Scholar] [CrossRef]
  58. Gupta, S.; Takebe, N.; Lorusso, P. Targeting the Hedgehog pathway in cancer. Ther. Adv. Med. Oncol. 2010, 2, 237–250. [Google Scholar] [CrossRef]
  59. Ming, J.E.; Nanni, L.; Muenke, M.; Meinecke, P.; Pierpont, M.E.M.; Robin, N.H.; Young, I.D.; Roessler, E.; Steinhaus, K.; Bocian, M.; et al. The Mutational Spectrum of the Sonic Hedgehog Gene in Holoprosencephaly: SHH Mutations Cause a Significant Proportion of Autosomal Dominant Holoprosencephaly. Hum. Mol. Genet. 1999, 8, 2479–2488. [Google Scholar] [CrossRef]
  60. Di Magliano, M.P.; Hebrok, M. Hedgehog signalling in cancer formation and maintenance. Nat. Rev. Cancer 2003, 3, 903–911. [Google Scholar] [CrossRef]
  61. Fujii, K.; Miyashita, T. Gorlin syndrome (nevoid basal cell carcinoma syndrome): Update and literature review. Pediatrics Int. 2014, 56, 667–674. [Google Scholar] [CrossRef]
  62. Sahebjam, S.; Siu, L.L.; Razak, A.A. The Utility of Hedgehog Signaling Pathway Inhibition for Cancer. Oncologist 2012, 17, 1090–1099. [Google Scholar] [CrossRef] [Green Version]
  63. Chahal, K.K.; Parle, M.; Abagyan, R. Hedgehog pathway and smoothened inhibitors in cancer therapies. Anti-Cancer Drugs 2018, 29, 387–401. [Google Scholar] [CrossRef]
  64. Skoda, A.M.; Simovic, D.; Karin, V.; Kardum, V.; Vranic, S.; Serman, L. The role of the Hedgehog signaling pathway in cancer: A comprehensive review. Bosn. J. Basic Med. Sci. 2018, 18, 8–20. [Google Scholar] [CrossRef]
  65. Epstein, E.H. Basal cell carcinomas: Attack of the hedgehog. Nat. Rev. Cancer 2008, 8, 743–754. [Google Scholar] [CrossRef]
  66. Ouhtit, A.; Nakazawa, H.; Armstrong, B.K.; Kricker, A.; Tan, E.; Yamasaki, H.; English, D.R. UV-radiation-specific p53 mutation frequency in normal skin as a predictor of risk of basal cell carcinoma. J. Natl. Cancer Inst. 1998, 90, 523–531. [Google Scholar] [CrossRef]
  67. Furth, N.; Aylon, Y.; Oren, M. p53 shades of Hippo. Cell Death Differ. 2018, 25, 81–92. [Google Scholar] [CrossRef]
  68. Mancuso, M.; Pazzaglia, S.; Tanori, M.; Hahn, H.; Merola, P.; Rebessi, S.; Atkinson, M.J.; Di Majo, V.; Covelli, V.; Saran, A. Basal Cell Carcinoma and Its Development: Insights from Radiation-Induced Tumors in Ptch1-Deficient Mice. Cancer Res. 2004, 64, 934–941. [Google Scholar] [CrossRef]
  69. Mercurio, L.; Albanesi, C.; Madonna, S. Recent Updates on the Involvement of PI3K/AKT/mTOR Molecular Cascade in the Pathogenesis of Hyperproliferative Skin Disorders. Front. Med. 2021, 8, 665647. [Google Scholar] [CrossRef]
  70. Chakrabarti, J.; Holokai, L.; Syu, L.; Steele, N.G.; Chang, J.; Wang, J.; Ahmed, S.; Dlugosz, A.; Zavros, Y. Hedgehog signaling induces PD-L1 expression and tumor cell proliferation in gastric cancer. Oncotarget 2018, 9, 37439–37457. [Google Scholar] [CrossRef]
  71. Lipson, E.J.; Lilo, M.T.; Ogurtsova, A.; Esandrio, J.; Xu, H.; Brothers, P.; Schollenberger, M.; Sharfman, W.H.; Taube, J.M. Basal cell carcinoma: PD-L1/PD-1 checkpoint expression and tumor regression after PD-1 blockade. J. ImmunoTherapy Cancer 2017, 5, 23. [Google Scholar] [CrossRef] [PubMed]
  72. Weiss, G.J.; Korn, R.L. Metastatic basal cell carcinoma in the era of hedgehog signaling pathway inhibitors. Cancer 2012, 118, 5310–5319. [Google Scholar] [CrossRef] [PubMed]
  73. Ozgediz, D.; Smith, E.B.; Zheng, J.; Otero, J.; Tabatabai, Z.L.; Corvera, C.U. Basal cell carcinoma does metastasize. Derm. Online J. 2008, 14, 5. [Google Scholar] [CrossRef]
  74. Von Domarus, H.; Stevens, P.J. Metastatic basal cell carcinoma. Report of five cases and review of 170 cases in the literature. J. Am. Acad. Dermatol. 1984, 10, 1043–1060. [Google Scholar] [CrossRef]
  75. Lee, S.T.; Welch, K.D.; Panter, K.E.; Gardner, D.R.; Garrossian, M.; Chang, C.-W.T. Cyclopamine: From Cyclops Lambs to Cancer Treatment. J. Agric. Food Chem. 2014, 62, 7355–7362. [Google Scholar] [CrossRef]
  76. Binns, W.; Shupe, J.L.; Keeler, R.F.; James, L.F. Chronologic evaluation of teratogenicity in sheep fed Veratrum californicum. J. Am. Vet. Med. Assoc. 1965, 147, 839–842. [Google Scholar]
  77. James, L.F. Teratological research at the USDA-ARS poisonous plant research laboratory. J. Nat. Toxins 1999, 8, 63–80. [Google Scholar]
  78. Chiang, C.; Litingtung, Y.; Lee, E.; Young, K.E.; Corden, J.L.; Westphal, H.; Beachy, P.A. Cyclopia and defective axial patterning in mice lacking Sonic hedgehog gene function. Nature 1996, 383, 407–413. [Google Scholar] [CrossRef]
  79. Litingtung, Y.; Lei, L.; Westphal, H.; Chiang, C. Sonic hedgehog is essential to foregut development. Nat. Genet. 1998, 20, 58–61. [Google Scholar] [CrossRef]
  80. Cooper, M.K.; Porter, J.A.; Young, K.E.; Beachy, P.A. Teratogen-mediated inhibition of target tissue response to Shh signaling. Science 1998, 280, 1603–1607. [Google Scholar] [CrossRef]
  81. Incardona, J.P.; Gaffield, W.; Kapur, R.P.; Roelink, H. The teratogenic Veratrum alkaloid cyclopamine inhibits sonic hedgehog signal transduction. Development 1998, 125, 3553–3562. [Google Scholar] [CrossRef]
  82. Sanchez, P.; i Altaba, A.R. In vivo inhibition of endogenous brain tumors through systemic interference of Hedgehog signaling in mice. Mech. Dev. 2005, 122, 223–230. [Google Scholar] [CrossRef]
  83. 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. Medulloblastoma growth inhibition by hedgehog pathway blockade. Science 2002, 297, 1559–1561. [Google Scholar] [CrossRef]
  84. Clement, V.; Sanchez, P.; de Tribolet, N.; Radovanovic, I.; Ruiz i Altaba, A. HEDGEHOG-GLI1 signaling regulates human glioma growth, cancer stem cell self-renewal, and tumorigenicity. Curr. Biol. 2007, 17, 165–172. [Google Scholar] [CrossRef]
  85. Stecca, B.; Mas, C.; Clement, V.; Zbinden, M.; Correa, R.; Piguet, V.; Beermann, F.; Ruiz i Altaba, A. Melanomas require HEDGEHOG-GLI signaling regulated by interactions between GLI1 and the RAS-MEK/AKT pathways. Proc. Natl. Acad. Sci. USA 2007, 104, 5895–5900. [Google Scholar] [CrossRef]
  86. 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]
  87. 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]
  88. Karhadkar, S.S.; Steven Bova, G.; 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]
  89. Frampton, J.E.; Basset-Séguin, N. Vismodegib: A Review in Advanced Basal Cell Carcinoma. Drugs 2018, 78, 1145–1156. [Google Scholar] [CrossRef]
  90. Jain, S.; Song, R.; Xie, J. Sonidegib: Mechanism of action, pharmacology, and clinical utility for advanced basal cell carcinomas. Onco. Targets Ther. 2017, 10, 1645–1653. [Google Scholar] [CrossRef]
  91. Chang, A.L.S.; Solomon, J.A.; Hainsworth, J.D.; Goldberg, L.; McKenna, E.; Day, B.-m.; Chen, D.M.; Weiss, G.J. Expanded access study of patients with advanced basal cell carcinoma treated with the Hedgehog pathway inhibitor, vismodegib. J. Am. Acad. Dermatol. 2014, 70, 60–69. [Google Scholar] [CrossRef] [PubMed]
  92. Chang, A.L.S.; Oro, A.E. Initial assessment of tumor regrowth after vismodegib in advanced basal cell carcinoma. Arch. Dermatol. 2012, 148, 1324–1325. [Google Scholar] [CrossRef] [PubMed]
  93. 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. Genomic analysis of smoothened inhibitor resistance in basal cell carcinoma. Cancer Cell 2015, 27, 327–341. [Google Scholar] [CrossRef] [PubMed]
  94. 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.S. Smoothened variants explain the majority of drug resistance in basal cell carcinoma. Cancer Cell 2015, 27, 342–353. [Google Scholar] [CrossRef] [Green Version]
  95. Ridky, T.W.; Cotsarelis, G. Vismodegib resistance in basal cell carcinoma: Not a smooth fit. Cancer Cell 2015, 27, 315–316. [Google Scholar] [CrossRef]
  96. Danial, C.; Sarin, K.Y.; Oro, A.E.; Chang, A.L.S. 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]
  97. Gutzmer, R.; Solomon, J.A. Hedgehog Pathway Inhibition for the Treatment of Basal Cell Carcinoma. Target. Oncol. 2019, 14, 253–267. [Google Scholar] [CrossRef]
  98. Li, Q.-r.; Zhao, H.; Zhang, X.-s.; Lang, H.; Yu, K. Novel-smoothened inhibitors for therapeutic targeting of naïve and drug-resistant hedgehog pathway-driven cancers. Acta Pharmacol. Sin. 2019, 40, 257–267. [Google Scholar] [CrossRef]
  99. Zárate, A.M.; Espinosa-Bustos, C.; Guerrero, S.; Fierro, A.; Oyarzún-Ampuero, F.; Quest, A.F.G.; Di Marcotullio, L.; Loricchio, E.; Caimano, M.; Calcaterra, A.; et al. A New Smoothened Antagonist Bearing the Purine Scaffold Shows Antitumour Activity In Vitro and In Vivo. Int. J. Mol. Sci. 2021, 22, 8372. [Google Scholar] [CrossRef]
  100. Hoch, L.; Faure, H.; Roudaut, H.; Schoenfelder, A.; Mann, A.; Girard, N.; Bihannic, L.; Ayrault, O.; Petricci, E.; Taddei, M.; et al. MRT-92 inhibits Hedgehog signaling by blocking overlapping binding sites in the transmembrane domain of the Smoothened receptor. FASEB J. 2015, 29, 1817–1829. [Google Scholar] [CrossRef]
  101. Pietrobono, S.; Santini, R.; Gagliardi, S.; Dapporto, F.; Colecchia, D.; Chiariello, M.; Leone, C.; Valoti, M.; Manetti, F.; Petricci, E.; et al. Targeted inhibition of Hedgehog-GLI signaling by novel acylguanidine derivatives inhibits melanoma cell growth by inducing replication stress and mitotic catastrophe. Cell Death Dis. 2018, 9, 142. [Google Scholar] [CrossRef]
  102. Švenda, J.; Sheremet, M.; Kremer, L.; Maier, L.; Bauer, J.O.; Strohmann, C.; Ziegler, S.; Kumar, K.; Waldmann, H. Biology-Oriented Synthesis of a Withanolide-Inspired Compound Collection Reveals Novel Modulators of Hedgehog Signaling. Angew. Chem. Int. Ed. 2015, 54, 5596–5602. [Google Scholar] [CrossRef]
  103. Bonandi, E.; Mori, M.; Infante, P.; Basili, I.; Di Marcotullio, L.; Calcaterra, A.; Catti, F.; Botta, B.; Passarella, D. Design and Synthesis of New Withaferin A Inspired Hedgehog Pathway Inhibitors. Chem.—Eur. J. 2021, 27, 8350–8357. [Google Scholar] [CrossRef]
  104. Biehs, B.; Dijkgraaf, G.J.P.; Piskol, R.; Alicke, B.; Boumahdi, S.; Peale, F.; Gould, S.E.; de Sauvage, F.J. A cell identity switch allows residual BCC to survive Hedgehog pathway inhibition. Nature 2018, 562, 429–433. [Google Scholar] [CrossRef]
  105. Whitson, R.J.; Lee, A.; Urman, N.M.; Mirza, A.; Yao, C.Y.; Brown, A.S.; Li, J.R.; Shankar, G.; Fry, M.A.; Atwood, S.X.; et al. Noncanonical hedgehog pathway activation through SRF-MKL1 promotes drug resistance in basal cell carcinomas. Nat. Med. 2018, 24, 271–281. [Google Scholar] [CrossRef]
  106. Infante, P.; Alfonsi, R.; Botta, B.; Mori, M.; Di Marcotullio, L. Targeting GLI factors to inhibit the Hedgehog pathway. Trends Pharmacol. Sci. 2015, 36, 547–558. [Google Scholar] [CrossRef]
  107. Hopkins, A.L.; Groom, C.R. The druggable genome. Nat. Rev. Drug Discov. 2002, 1, 727–730. [Google Scholar] [CrossRef]
  108. Overington, J.P.; Al-Lazikani, B.; Hopkins, A.L. How many drug targets are there? Nat. Rev. Drug Discov. 2006, 5, 993–996. [Google Scholar] [CrossRef]
  109. Imming, P.; Sinning, C.; Meyer, A. Drugs, their targets and the nature and number of drug targets. Nat. Rev. Drug Discov. 2006, 5, 821–834. [Google Scholar] [CrossRef]
  110. Kim, J.; Lee, J.J.; Kim, J.; Gardner, D.; Beachy, P.A. Arsenic antagonizes the Hedgehog pathway by preventing ciliary accumulation and reducing stability of the Gli2 transcriptional effector. Proc. Natl. Acad. Sci. USA 2010, 107, 13432–13437. [Google Scholar] [CrossRef]
  111. Kim, J.; Aftab, B.; Tang, J.; Kim, D.; Lee, A.; Rezaee, M.; Kim, J.; Chen, B.; King, E.; Borodovsky, A.; et al. Itraconazole and Arsenic Trioxide Inhibit Hedgehog Pathway Activation and Tumor Growth Associated with Acquired Resistance to Smoothened Antagonists. Cancer Cell 2013, 23, 23–34. [Google Scholar] [CrossRef]
  112. Miller, W.H., Jr. Molecular Targets of Arsenic Trioxide in Malignant Cells. Oncologist 2002, 7 (Suppl. S1), 14–19. [Google Scholar] [CrossRef]
  113. Soignet, S.L.; Maslak, P.; Wang, Z.-G.; Jhanwar, S.; Calleja, E.; Dardashti, L.J.; Corso, D.; DeBlasio, A.; Gabrilove, J.; Scheinberg, D.A.; et al. Complete Remission after Treatment of Acute Promyelocytic Leukemia with Arsenic Trioxide. N. Engl. J. Med. 1998, 339, 1341–1348. [Google Scholar] [CrossRef]
  114. Cohen, M.H.; Hirschfeld, S.; Honig, S.F.; Ibrahim, A.; Johnson, J.R.; O’Leary, J.J.; White, R.M.; Williams, G.A.; Pazdur, R. Drug Approval Summaries: Arsenic Trioxide, Tamoxifen Citrate, Anastrazole, Paclitaxel, Bexarotene. Oncologist 2001, 6, 4–11. [Google Scholar] [CrossRef]
  115. Wang, Y.; Han, C.; Lu, L.; Magliato, S.; Wu, T. Hedgehog signaling pathway regulates autophagy in human hepatocellular carcinoma cells. Hepatology 2013, 58, 995–1010. [Google Scholar] [CrossRef]
  116. Chen, Q.; Xu, R.; Zeng, C.; Lu, Q.; Huang, D.; Shi, C.; Zhang, W.; Deng, L.; Yan, R.; Rao, H. Down-regulation of Gli transcription factor leads to the inhibition of migration and invasion of ovarian cancer cells via integrin β4-mediated FAK signaling. PLoS ONE 2014, 9, e88386. [Google Scholar] [CrossRef]
  117. Kebenko, M.; Drenckhan, A.; Gros, S.J.; Jücker, M.; Grabinski, N.; Ewald, F.; Grottke, A.; Schultze, A.; Izbicki, J.R.; Bokemeyer, C. ErbB2 signaling activates the Hedgehog pathway via PI3K–Akt in human esophageal adenocarcinoma: Identification of novel targets for concerted therapy concepts. Cell. Signal. 2015, 27, 373–381. [Google Scholar] [CrossRef]
  118. Santini, R.; Vinci, M.C.; Pandolfi, S.; Penachioni, J.Y.; Montagnani, V.; Olivito, B.; Gattai, R.; Pimpinelli, N.; Gerlini, G.; Borgognoni, L. Hedgehog-GLI signaling drives self-renewal and tumorigenicity of human melanoma-initiating cells. Stem Cells 2012, 30, 1808–1818. [Google Scholar] [CrossRef]
  119. 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]
  120. Lauth, M.; Bergstrm, s.; Shimokawa, T.; Toftgrd, 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]
  121. Huang, L.; Walter, V.; Hayes, D.N.; Onaitis, M. Hedgehog–GLI signaling inhibition suppresses tumor growth in squamous lung cancer. Clin. Cancer Res. 2014, 20, 1566–1575. [Google Scholar] [CrossRef] [PubMed]
  122. 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]
  123. Kiran Riaz, S.; Ke, Y.; Wang, F.; Kayani, M.; Malik, F. Influence of SHH/GLI1 axis on EMT mediated migration and invasion of breast cancer cells. Sci. Rep. 2019, 9, 6620. [Google Scholar] [CrossRef] [PubMed]
  124. Geng, L.; Zhou, X.; Li, X.; Lu, K.; Li, P.; Wang, X. Gli1 Inhibitor GANT61 Exhibits Antitumor Efficacy in T-Cell Lymphoma Cells through Down-Regulation of p-STAT3 and SOCS3 Pathways. Blood 2015, 126, 1287. [Google Scholar] [CrossRef]
  125. Calcaterra, A.; Iovine, V.; Botta, B.; Quaglio, D.; D’Acquarica, I.; Ciogli, A.; Iazzetti, A.; Alfonsi, R.; Lospinoso Severini, L.; Infante, P.; et al. Chemical, computational and functional insights into the chemical stability of the Hedgehog pathway inhibitor GANT61. J. Enzym. Inhib. Med. Chem. 2018, 33, 349–358. [Google Scholar] [CrossRef]
  126. Agyeman, A.; Jha, B.K.; Mazumdar, T.; Houghton, J.A. Mode and specificity of binding of the small molecule GANT61 to GLI determines inhibition of GLI-DNA binding. Oncotarget 2014, 5, 4492–4503. [Google Scholar] [CrossRef]
  127. Zhang, R.; Wu, J.; Ferrandon, S.; Glowacki, K.J.; Houghton, J.A. Targeting GLI by GANT61 involves mechanisms dependent on inhibition of both transcription and DNA licensing. Oncotarget 2016, 7, 80190–80207. [Google Scholar] [CrossRef]
  128. Borah, A.; Palaninathan, V.; Girija, A.R.; Balasubramanian, S.; Rochani, A.K. Poly-lactic-co-glycolic acid Nanoformulation of Small Molecule Antagonist GANT61 for Cancer Annihilation by Modulating Hedgehog Pathway. Nano World J. 2017, 3, 10. [Google Scholar] [CrossRef]
  129. Infante, P.; Mori, M.; Alfonsi, R.; Ghirga, F.; Aiello, F.; Toscano, S.; Ingallina, C.; Siler, M.; Cucchi, D.; Po, A.; et al. Gli1/DNA interaction is a druggable target for Hedgehog-dependent tumors. EMBO J. 2015, 34, 200–217. [Google Scholar] [CrossRef]
  130. Infante, P.; Malfanti, A.; Quaglio, D.; Balducci, S.; De Martin, S.; Bufalieri, F.; Mastrotto, F.; Basili, I.; Garofalo, M.; Lospinoso Severini, L.; et al. Glabrescione B delivery by self-assembling micelles efficiently inhibits tumor growth in preclinical models of Hedgehog-dependent medulloblastoma. Cancer Lett. 2021, 499, 220–231. [Google Scholar] [CrossRef]
  131. Berardozzi, S.; Bernardi, F.; Infante, P.; Ingallina, C.; Toscano, S.; De Paolis, E.; Alfonsi, R.; Caimano, M.; Botta, B.; Mori, M.; et al. Synergistic inhibition of the Hedgehog pathway by newly designed Smo and Gli antagonists bearing the isoflavone scaffold. Eur. J. Med. Chem. 2018, 156, 554–562. [Google Scholar] [CrossRef]
  132. Lospinoso Severini, L.; Quaglio, D.; Basili, I.; Ghirga, F.; Bufalieri, F.; Caimano, M.; Balducci, S.; Moretti, M.; Romeo, I.; Loricchio, E.; et al. A Smo/Gli Multitarget Hedgehog Pathway Inhibitor Impairs Tumor Growth. Cancers 2019, 11, 1518. [Google Scholar] [CrossRef]
  133. Hom, M.E.; Ondrus, A.E.; Sakata-Kato, T.; Rack, P.G.; Chen, J.K. Bicyclic Imidazolium Inhibitors of Gli Transcription Factor Activity. ChemMedChem 2020, 15, 1044–1049. [Google Scholar] [CrossRef]
  134. Gómez-Ruiz, S.; Maksimović-Ivanić, D.; Mijatović, S.; Kaluđerović, G.N. On the discovery, biological effects, and use of Cisplatin and metallocenes in anticancer chemotherapy. Bioinorg. Chem. Appl. 2012, 2012, 140284. [Google Scholar] [CrossRef] [Green Version]
  135. Orvig, C.; Abrams, M.J. Medicinal Inorganic Chemistry:  Introduction. Chem. Rev. 1999, 99, 2201–2204. [Google Scholar] [CrossRef]
  136. Dukes, M.W.; Bajema, E.A.; Whittemore, T.J.; Holmgren, R.A.; Meade, T.J. Delivery of Targeted Co(III)–DNA Inhibitors of Gli Proteins to Disrupt Hedgehog Signaling. Bioconjug. Chem. 2022, 33, 643–653. [Google Scholar] [CrossRef]
  137. Brue, C.R.; Dukes, M.W.; Masotti, M.; Holmgren, R.; Meade, T.J. Functional Disruption of Gli1-DNA Recognition via a Cobalt(III) Complex. ChemMedChem 2022, 17, e202200025. [Google Scholar] [CrossRef]
  138. Manus, L.M.; Holbrook, R.J.; Atesin, T.A.; Heffern, M.C.; Harney, A.S.; Eckermann, A.L.; Meade, T.J. Axial Ligand Exchange of N-heterocyclic Cobalt(III) Schiff Base Complexes: Molecular Structure and NMR Solution Dynamics. Inorg. Chem. 2013, 52, 1069–1076. [Google Scholar] [CrossRef]
  139. Harney, A.S.; Lee, J.; Manus, L.M.; Wang, P.; Ballweg, D.M.; LaBonne, C.; Meade, T.J. Targeted inhibition of Snail family zinc finger transcription factors by oligonucleotide-Co(III) Schiff base conjugate. Proc. Natl. Acad. Sci. USA 2009, 106, 13667–13672. [Google Scholar] [CrossRef]
  140. Harney, A.S.; Meade, T.J.; LaBonne, C. Targeted Inactivation of Snail Family EMT Regulatory Factors by a Co(III)-Ebox Conjugate. PLoS ONE 2012, 7, e32318. [Google Scholar] [CrossRef]
  141. Hurtado, R.R.; Harney, A.S.; Heffern, M.C.; Holbrook, R.J.; Holmgren, R.A.; Meade, T.J. Specific Inhibition of the Transcription Factor Ci by a Cobalt(III) Schiff Base–DNA Conjugate. Mol. Pharm. 2012, 9, 325–333. [Google Scholar] [CrossRef]
  142. Sasaki, H.; Nishizaki, Y.; Hui, C.-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] [CrossRef]
  143. Yun, T.; Wang, J.; Yang, J.; Huang, W.; Lai, L.; Tan, W.; Liu, Y. Discovery of Small Molecule Inhibitors Targeting the Sonic Hedgehog. Front. Chem. 2020, 8, 498. [Google Scholar] [CrossRef]
  144. Riobó, N.A.; Lu, K.; Ai, X.; Haines, G.M.; Emerson, C.P., Jr. Phosphoinositide 3-kinase and Akt are essential for Sonic Hedgehog signaling. Proc. Natl. Acad. Sci. USA 2006, 103, 4505–4510. [Google Scholar] [CrossRef] [Green Version]
  145. Gordon, R.E.; Zhang, L.; Peri, S.; Kuo, Y.-M.; Du, F.; Egleston, B.L.; Ng, J.M.Y.; Andrews, A.J.; Astsaturov, I.; Curran, T.; et al. Statins Synergize with Hedgehog Pathway Inhibitors for Treatment of Medulloblastoma. Clin. Cancer Res. 2018, 24, 1375–1388. [Google Scholar] [CrossRef]
  146. Bijlsma, M.F.; Spek, C.A.; Zivkovic, D.; van de Water, S.; Rezaee, F.; Peppelenbosch, M.P. Repression of Smoothened by Patched-Dependent (Pro-)Vitamin D3 Secretion. PLoS Biol. 2006, 4, e232. [Google Scholar] [CrossRef]
  147. Kinto, N.; Iwamoto, M.; Enomoto-Iwamoto, M.; Noji, S.; Ohuchi, H.; Yoshioka, H.; Kataoka, H.; Wada, Y.; Yuhao, G.; Takahashi, H.E.; et al. Fibroblasts expressing Sonic hedgehog induce osteoblast differentiation and ectopic bone formation. FEBS Lett. 1997, 404, 319–323. [Google Scholar] [CrossRef]
  148. Nakamura, T.; Aikawa, T.; Iwamoto-Enomoto, M.; Iwamoto, M.; Higuchi, Y.; Maurizio, P.; Kinto, N.; Yamaguchi, A.; Noji, S.; Kurisu, K.; et al. Induction of Osteogenic Differentiation by Hedgehog Proteins. Biochem. Biophys. Res. Commun. 1997, 237, 465–469. [Google Scholar] [CrossRef]
  149. Spinella-Jaegle, S.; Rawadi, G.; Kawai, S.; Gallea, S.; Faucheu, C.; Mollat, P.; Courtois, B.; Bergaud, B.; Ramez, V.; Blanchet, A.M.; et al. Sonic hedgehog increases the commitment of pluripotent mesenchymal cells into the osteoblastic lineage and abolishes adipocytic differentiation. J. Cell Sci. 2001, 114 Pt 11, 2085–2094. [Google Scholar] [CrossRef]
  150. Ingram, W.J.; Wicking, C.A.; Grimmond, S.M.; Forrest, A.R.; Wainwright, B.J. Novel genes regulated by Sonic Hedgehog in pluripotent mesenchymal cells. Oncogene 2002, 21, 8196–8205. [Google Scholar] [CrossRef]
  151. Oro, A.E.; Higgins, K.M.; Hu, Z.; Bonifas, J.M.; Epstein, E.H.; Scott, M.P. Basal Cell Carcinomas in Mice Overexpressing Sonic Hedgehog. Science 1997, 276, 817–821. [Google Scholar] [CrossRef] [PubMed]
  152. 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]
  153. Aszterbaum, M.; Epstein, J.; Oro, A.; Douglas, V.; LeBoit, P.E.; Scott, M.P.; Epstein, E.H. 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]
  154. Nitzki, F.; Becker, M.; Frommhold, A.; Schulz-Schaeffer, W.; Hahn, H. Patched knockout mouse models of Basal cell carcinoma. J. Ski. Cancer 2012, 2012, 907543. [Google Scholar] [CrossRef] [Green Version]
  155. Goodrich Lisa, V.; Milenković, L.; Higgins Kay, M.; Scott Matthew, P. Altered Neural Cell Fates and Medulloblastoma in Mouse patched Mutants. Science 1997, 277, 1109–1113. [Google Scholar] [CrossRef]
  156. Hahn, H.; Wojnowski, L.; Zimmer, A.M.; Hall, J.; Miller, G.; Zimmer, A. Rhabdomyosarcomas and radiation hypersensitivity in a mouse model of Gorlin syndrome. Nat. Med. 1998, 4, 619–622. [Google Scholar] [CrossRef]
  157. Collins, A.T.; Lang, S.H. A systematic review of the validity of patient derived xenograft (PDX) models: The implications for translational research and personalised medicine. PeerJ 2018, 6, e5981. [Google Scholar] [CrossRef]
  158. Pawlowski, A.; Haberman, H.F. Heterotransplantation of human basal cell carcinomas in “nude” mice. J. Investig. Dermatol. 1979, 72, 310–313. [Google Scholar] [CrossRef]
  159. Grimwood, R.E.; Johnson, C.A.; Ferris, C.F.; Mercill, D.B.; Mellette, J.R.; Huff, J.C. Transplantation of human basal cell carcinomas to athymic mice. Cancer 1985, 56, 519–523. [Google Scholar] [CrossRef]
  160. Grimwood, R.E.; Tharp, M.D. Growth of Human Basal Cell Carcinomas Transplanted to C57/Balb/C bgJ/bgJ-nu/nu (Beige-Nude) Mice. J. Dermatol. Surg. Oncol. 1991, 17, 661–666. [Google Scholar] [CrossRef]
  161. Carlson, J.A.; Combates, N.J.; Stenn, K.S.; Prouty, S.M. Anaplastic neoplasms arising from basal cellcarcinoma xenotransplants into SCID-beige mice. J. Cutan. Pathol. 2002, 29, 268–278. [Google Scholar] [CrossRef]
  162. Wang, G.Y.; So, P.-L.; Wang, L.; Libove, E.; Wang, J.; Epstein, E.H. Establishment of Murine Basal Cell Carcinoma Allografts: A Potential Model for Preclinical Drug Testing and for Molecular Analysis. J. Investig. Dermatol. 2011, 131, 2298–2305. [Google Scholar] [CrossRef]
  163. Jiang, L.P.; Shen, Q.S.; Yang, C.P.; Chen, Y.B. Establishment of basal cell carcinoma animal model in Chinese tree shrew (Tupaia belangeri chinensis). Zool. Res. 2017, 38, 180–190. [Google Scholar] [CrossRef]
  164. Xu, L.; Zhang, Y.; Liang, B.; Lü, L.B.; Chen, C.S.; Chen, Y.B.; Zhou, J.M.; Yao, Y.G. Tree shrews under the spot light: Emerging model of human diseases. Dongwuxue Yanjiu 2013, 34, 59–69. [Google Scholar] [CrossRef]
  165. Zhang, J.; Luo, R.C.; Man, X.Y.; Lv, L.B.; Yao, Y.G.; Zheng, M. The anatomy of the skin of the Chinese tree shrew is very similar to that of human skin. Zool. Res. 2020, 41, 208. [Google Scholar] [CrossRef]
  166. Gaviria Agudelo, C.; Restrepo, L.M. Human Skin Cancer: An Overview of Animal, Ex Vivo, and In Vitro Models. Curr. Dermatol. Rep. 2022, 11, 168–177. [Google Scholar] [CrossRef]
Scheme 1. Cartoon representation of epidermal layers and the growth of BCC. Tumor cells originate from the stratum basale where they maintain stemness and hyperproliferative capacity. As the tumors progress, they begin to spread and migrate into the dermis as well as alter the pathology of non-tumor keratinocyte differentiation.
Scheme 1. Cartoon representation of epidermal layers and the growth of BCC. Tumor cells originate from the stratum basale where they maintain stemness and hyperproliferative capacity. As the tumors progress, they begin to spread and migrate into the dermis as well as alter the pathology of non-tumor keratinocyte differentiation.
Biomedicines 10 02376 sch001
Scheme 2. Mechanism of PDT for cellular death. A photosensitizer is delivered to tumor tissue and irradiated with laser light that matches its excitation wavelength. Upon relaxation, energetic crossing transfers the energy to oxygen and water, generating cytotoxic singlet oxygen (1O2) and hydrogen peroxide (H2O2). Cell death is only observed where the laser light is administered, limiting damage to nearby healthy tissue.
Scheme 2. Mechanism of PDT for cellular death. A photosensitizer is delivered to tumor tissue and irradiated with laser light that matches its excitation wavelength. Upon relaxation, energetic crossing transfers the energy to oxygen and water, generating cytotoxic singlet oxygen (1O2) and hydrogen peroxide (H2O2). Cell death is only observed where the laser light is administered, limiting damage to nearby healthy tissue.
Biomedicines 10 02376 sch002
Figure 1. Chemical structures of imiquimod (left) and 5-florouracil (right). Imiquimod functions through an immunomodulatory response, and 5-florouracil inhibits DNA synthesis machinery. These treatments are effective topically against low-risk, superficial BCC.
Figure 1. Chemical structures of imiquimod (left) and 5-florouracil (right). Imiquimod functions through an immunomodulatory response, and 5-florouracil inhibits DNA synthesis machinery. These treatments are effective topically against low-risk, superficial BCC.
Biomedicines 10 02376 g001
Figure 2. Key regulators of the Hedgehog signaling cascade. 1. Hh signaling is activated by the binding of Hh ligands (Sonic Hedgehog, Indian Hedgehog, and Desert Hedgehog) to the transmembrane protein PTCH1. 2. PTCH1 is a suppressor of SMO activity. Upon the binding of Hh proteins, inhibition of SMO is released, represented by the red X. 3. Upon activation, SMO signals for the SUFU complex to release the Gli family of transcription factors. 4. Gli transcription factors are activated in the cytosol prior to translocating into the nucleus. 5. Gli transcription factors transcribe pro-proliferative and migratory genes that lead to tumorigenesis in BCC. Crystal structures images were made from the following Protein Data Bank files: Ptch1/SHH complex, 6N7H. SMO, 6D35. SUFU/Gli complex: 4BLB. Gli1/DNA complex, 2Gli.
Figure 2. Key regulators of the Hedgehog signaling cascade. 1. Hh signaling is activated by the binding of Hh ligands (Sonic Hedgehog, Indian Hedgehog, and Desert Hedgehog) to the transmembrane protein PTCH1. 2. PTCH1 is a suppressor of SMO activity. Upon the binding of Hh proteins, inhibition of SMO is released, represented by the red X. 3. Upon activation, SMO signals for the SUFU complex to release the Gli family of transcription factors. 4. Gli transcription factors are activated in the cytosol prior to translocating into the nucleus. 5. Gli transcription factors transcribe pro-proliferative and migratory genes that lead to tumorigenesis in BCC. Crystal structures images were made from the following Protein Data Bank files: Ptch1/SHH complex, 6N7H. SMO, 6D35. SUFU/Gli complex: 4BLB. Gli1/DNA complex, 2Gli.
Biomedicines 10 02376 g002
Figure 4. Chemical structures of withaferin A derivatives, novel SMO inhibitors that structurally deviate from Vismodegib. (A) Chemical structure of natural product withaferin A. The highlighted scaffold is believed to be important for SMO binding. (B,C) The Waldman group developed derivatives of withaferin A that exhibited stereoselective potent inhibition of SMO. (D) The target compound of the Passarella group for SMO inhibition did not inhibit the Hh pathway. * represent asymmetric carbons. (E,F) Synthetic intermediates 13b and 14b exhibited stereoselective inhibition of SMO, elucidating important polarity considerations for protein interactions.
Figure 4. Chemical structures of withaferin A derivatives, novel SMO inhibitors that structurally deviate from Vismodegib. (A) Chemical structure of natural product withaferin A. The highlighted scaffold is believed to be important for SMO binding. (B,C) The Waldman group developed derivatives of withaferin A that exhibited stereoselective potent inhibition of SMO. (D) The target compound of the Passarella group for SMO inhibition did not inhibit the Hh pathway. * represent asymmetric carbons. (E,F) Synthetic intermediates 13b and 14b exhibited stereoselective inhibition of SMO, elucidating important polarity considerations for protein interactions.
Biomedicines 10 02376 g004
Figure 5. Chemical structures for small molecule Gli inhibitors. (A) Arsenic trioxide downregulates Gli activity but is unknown to bind directly. (B) GANT-61 is the gold standard for direct Gli inhibition. In aqueous solution, it undergoes hydrolysis to unmask the active inhibiting scaffold. (C) Glabrescione B binds directly to Gli in disrupt the DNA binding interaction. (D) Structural modifications to GlaB elucidate chemical modifications that maintain or prevent Gli inhibition (E) A dual-targeted compound that inhibits both SMO and Gli. (F) New bicyclic imidazolium derivatives inhibit Gli function but have unintended impacts on cellular health.
Figure 5. Chemical structures for small molecule Gli inhibitors. (A) Arsenic trioxide downregulates Gli activity but is unknown to bind directly. (B) GANT-61 is the gold standard for direct Gli inhibition. In aqueous solution, it undergoes hydrolysis to unmask the active inhibiting scaffold. (C) Glabrescione B binds directly to Gli in disrupt the DNA binding interaction. (D) Structural modifications to GlaB elucidate chemical modifications that maintain or prevent Gli inhibition (E) A dual-targeted compound that inhibits both SMO and Gli. (F) New bicyclic imidazolium derivatives inhibit Gli function but have unintended impacts on cellular health.
Biomedicines 10 02376 g005
Figure 6. An inorganic inhibition approach for Gli. (A) Structure of the Gli binding domain co-crystalized with its native DNA binding partner (structure produced from PDB entry 2Gli). Inlet shows Cys2His2 zinc(II) coordination site that locks alpha-helical secondary structure into place. (B) Structure of CoGli, a cobalt-based inhibitor of Gli transcription factors. The Gli targeting sequence imparts specificity for Gli proteins and the cobalt complex displaces zinc ions. Evaluation of this complex in 2D BCC assays produced 50% inhibition of Hh-driven cell migration at 300 nM compared to 5 µM GANT-61. * Denotes phosphorothioate linkages to evade degradation by endonucleases.
Figure 6. An inorganic inhibition approach for Gli. (A) Structure of the Gli binding domain co-crystalized with its native DNA binding partner (structure produced from PDB entry 2Gli). Inlet shows Cys2His2 zinc(II) coordination site that locks alpha-helical secondary structure into place. (B) Structure of CoGli, a cobalt-based inhibitor of Gli transcription factors. The Gli targeting sequence imparts specificity for Gli proteins and the cobalt complex displaces zinc ions. Evaluation of this complex in 2D BCC assays produced 50% inhibition of Hh-driven cell migration at 300 nM compared to 5 µM GANT-61. * Denotes phosphorothioate linkages to evade degradation by endonucleases.
Biomedicines 10 02376 g006
Table 1. Select list and commentary of treatment options for BCC.
Table 1. Select list and commentary of treatment options for BCC.
TreatmentAdvantagesDisadvantagesPatient Restrictions
Wide Local Excision (WLE)Low recurrence rates upon complete excision

Short procedure times
Excision likely to be
incomplete and lead to higher recurrence rate

Highly invasive
Elderly patients where surgery is considered risky

When loss of anatomical
function is a risk
Mohs Micrographic Surgery (MMS)Promotes excision of poorly defined tumor margins

Suggested for more
aggressive/high-risk tumors

Minimizes harm to
non-diseased tissue
Long treatment times

Requires highly trained
physicians, and access may
be limited to patients in
underdeveloped areas
Elderly patients where surgery is considered risky

When loss of anatomical
function is a risk
Radiation TherapyCan treat tumors in locations where the loss of anatomical function is a risk

Can be used in higher-risk BCC

Boosts the efficacy of
incomplete surgical resection when used in tandem
Use of ionizing radiation
Increases risk of melanoma

Not as effective in larger
Tumors

Cannot determine the
complete clearance of tumor tissue
Not advised for patients with Gorlin Syndrome

Not advised for younger
patients due to the long-term impact of ionizing radiation
Ablative Laser
Therapy
Locally delivered, less
destructive to non-diseased
tissue

Low recurrence rates

Favorable cosmetic outcomes
Not applicable to larger, deeper tissues

May induce increased
sensitivity to the sun
Not recommended for
patients with high sunlight
sensitivity

Not applicable for patients
with nodular or infiltrative
BCC
Photodynamic
Therapy (PDT)
Well characterized
mechanisms of cytotoxicity

Local administration of
non-harmful laser light

Safe for patients with Gorlin Syndrome
High variability of
treatment efficacy

Multiple treatment sessions

Increased sensitivity to
Sunlight

Severely limited depth
penetration
Not recommended for
patients with high sunlight
sensitivity

Not applicable for patients
with nodular or infiltrative
BCC
Imiquimod
Topical Therapy
Well-characterized induction of immune response

Topical, localized
application reduces harm to healthy tissue
Only approved for small
superficial BCCs

Many patients report skin
Irritation

Recurrence rates understudied
Not applicable for patients
with nodular or infiltrative
BCC
5-Fluorouracil
Topical Therapy
Well-characterized inhibition of DNA synthesis

Topical, localized application reduces harm to healthy tissue

High cure rate
Not specific to tumor tissue and may cause harm to non-diseased skin

Many patients report skin
Irritation

Recurrence rates understudied
Not applicable for patients with nodular or infiltrative BCC
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Dukes, M.W.; Meade, T.J. Modulation of Hedgehog Signaling for the Treatment of Basal Cell Carcinoma and the Development of Preclinical Models. Biomedicines 2022, 10, 2376. https://doi.org/10.3390/biomedicines10102376

AMA Style

Dukes MW, Meade TJ. Modulation of Hedgehog Signaling for the Treatment of Basal Cell Carcinoma and the Development of Preclinical Models. Biomedicines. 2022; 10(10):2376. https://doi.org/10.3390/biomedicines10102376

Chicago/Turabian Style

Dukes, Meghan W., and Thomas J. Meade. 2022. "Modulation of Hedgehog Signaling for the Treatment of Basal Cell Carcinoma and the Development of Preclinical Models" Biomedicines 10, no. 10: 2376. https://doi.org/10.3390/biomedicines10102376

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