Next Article in Journal
Trimodal Therapy in Esophageal Squamous Cell Carcinoma: Role of Adjuvant Therapy Following Neoadjuvant Chemoradiation and Surgery
Next Article in Special Issue
Feasibility of Intratumoral Anti-PD1 as Treatment of Human Basal Cell Carcinoma: An Explorative Study with Adjuvant Ablative Fractional Laser
Previous Article in Journal
Protein Panel of Serum-Derived Small Extracellular Vesicles for the Screening and Diagnosis of Epithelial Ovarian Cancer
Previous Article in Special Issue
Advances in Cutaneous Squamous Cell Carcinoma Management
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advances in Management and Therapeutics of Cutaneous Basal Cell Carcinoma

1
Department of Dermatology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
2
Department of Dermatology, McGovern Medical School, University of Texas, Houston, TX 77030, USA
3
Department of Dermatology and Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(15), 3720; https://doi.org/10.3390/cancers14153720
Submission received: 1 July 2022 / Revised: 27 July 2022 / Accepted: 27 July 2022 / Published: 30 July 2022
(This article belongs to the Special Issue Research Progress of Cutaneous Squamous and Basal Cell Carcinomas)

Abstract

:

Simple Summary

Basal cell carcinoma (BCC) is the most common malignancy in humans with a range of treatment options available. Tumor and patient characteristics aid in risk-stratification, which influences treatment considerations. Here, we review the advancements in surgical, topical, field, immunotherapeutic, molecular-targeted, and experimental treatment modalities that can be employed in the correct clinical setting for the treatment of BCC.

Abstract

Basal cell carcinoma (BCC), the most common cancer in humans, is a malignant neoplasm of cells derived from the basal layer of the epidermis. Tumor characteristics such as histologic subtype, primary versus recurrent tumor, anatomic location, size, and patient attributes determine the risk level and acceptable treatment options. Surgical options offer histologic confirmation of tumor clearance. Standard excision provides post-treatment histologic assessment, while Mohs micrographic surgery (MMS) provides complete margin assessment intraoperatively. Additional treatment options may be employed in the correct clinical context. Small and low-risk BCCs, broad field cancerization, locally-advanced disease, metastatic disease, cosmetic concerns, or morbidity with surgical approaches raise consideration of other treatment modalities. We review herein a range of treatment approaches and advances in treatments for BCC, including standard excision, MMS, electrodesiccation and curettage, ablative laser treatment, radiation therapy, targeted molecular therapies, topical therapies, field therapies, immunotherapy, and experimental therapies.

1. Introduction

Non-melanoma skin cancers (NMSCs) represent approximately one-third of all malignancies in the United States [1]. Basal cell carcinoma (BCC) incidence worldwide has risen steadily by 3–10% each year with treatment of BCC in the 2013 U.S. Medicare population accounting for over $700 million in healthcare expenditures [2,3].Precise epidemiological data on BCC incidence is limited because NMSC is not reliably recorded by cancer registries. Factors conferring an increased risk of BCC development include ultraviolet radiation exposure, fair complexion, immunosuppression, advanced age, and genetic predisposing conditions (i.e., basal cell nevus syndrome (BCNS), xeroderma pigmentosum) [4]. Prognosis is generally excellent for well-defined, small, slow-growing BCCs in low-risk areas, which can be managed with surgical interventions, destructive localized treatments, and non-destructive topical and field therapies [5]. However, a subset of BCCs demonstrate features that predict an aggressive course (see Supplementary Table S1). Some of these features include large size, high-risk histologic subtype (i.e., sclerosing, micronodular, morpheaform, infiltrative, and basosquamous growth patterns), systemic drug resistance, recurrent disease, locally-advanced BCC (laBCC) or metastatic BCC (mBCC). Together, they cause significant morbidity and non-negligible mortality [5,6]. The heterogeneity in behavior of BCCs points to diverse underlying genetic drivers and the need for varied and targeted tools in the treatment paradigm.
Early studies of basal cell nevus syndrome revealed aberrant signaling in the Sonic Hedgehog (Hh) pathway characterized by an inactivating germline mutation in the Hh receptor, Patched (PTCH1) [7]. Resultant disinhibition of the transmembrane protein Smoothened (SMO) activates the glioma-associated oncogene (GLI) transcription factors and gene expression governing cell growth and proliferation. Sporadic BCCs also harbor Hh pathway mutations in PTCH1, SMO, and the negative GLI regulator suppressor of fused (SUFU) [7]. A recent genomic analysis of BCCs, revealed Hh pathway mutations in 85% of BCCs; however, the same proportion of BCCs harbored additional mutations in other major cancer-related pathways such as N-Myc (30% of BCCs) and Hippo-YAP (PTPN14 23% and LATS1 8%) [8]. These pathways underscore the diverse and complex underlying molecular drivers of oncogenesis in BCC and are potential therapeutic targets. Here, we review the advances in the management and therapeutic options for BCC treatment (see Table 1).

2. Procedural Treatments

2.1. Standard Excision with Postoperative Margin Assessment

The ideal treatment for most BCCs continues to be complete surgical removal [9]. In 1987, Wolf and Zitelli published recommendations on the surgical margins required for the surgical excision of BCCs [10]. Well-circumscribed BCCs smaller than 2-cm in diameter excised with a 4-mm margin had 98% clearance at 5-years. Margins of 2 or 3-mm eliminated 75 to 85% [10,11,12,13,14]. In 2020, Quazi et al. conducted a systematic review to identify necessary surgical margins on a larger sample size [15]. Well-demarcated BCCs smaller than 2-cm excised with 5 mm, 4 mm, 3 mm, and 2 mm margins showed complete excision rates of 94.73%, 92.22%, 90.34%, and 88.15%, respectively. Although the depth of invasion was not quantified, depth of recommended excision is the mid-subcutaneous tissue, or to the fascia, perichondrium, or periosteum if the subcutaneous layer is minimal [10]. For low-risk primary BCCs on the trunk and extremities, (excluding hands, feet, and the pretibial area) with positive postoperative margins, re-excision is indicated [10]. Because margin assessment is postoperative, recommendations for closure include secondary intent or linear closure, rather than tissue rearrangement [16]. Given the high risk of complication following second intention closures and the difficulty in attaining clear margins in larger lesions, each case must be evaluated by a team of specialists [17]. High-risk BCCs, such as the aggressive infiltrative variant which displays an increased recurrence risk, require a more careful approach with accurate and complete evaluation of surgical margins [18]. For more complex, high-risk recurrent tumors, and those on cosmetically sensitive sites, Mohs micrographic surgery is recommended [16].

2.2. Mohs Micrographic Surgery

Standard surgical excisions have recommended guidelines on appropriate surgical margins for low-risk BCCs on low-risk anatomic locations [16]. In contrast, higher-risk BCCs in higher-risk anatomic locations are more suitable for Mohs micrographic surgery (MMS), a technique in which tumors are excised with narrow surgical margins and intraoperative, frozen-section histological examination of the entire peripheral and deep margins is performed by the surgeon [16]. Residual tumor identified is excised by the same process, which is repeated until margins are tumor-free. MMS is a tissue sparing technique with lower recurrence rates compared to standard surgical excision [12,19,20]. According to a case series by Frederick Mohs and Anthony Emmett, cancers treated by MMS have recurrence rates around 1–2% with MMS demonstrating the lead cure rate for operable BCCs [19]. Two meta-analyses found the 5-year recurrence rate following MMS for primary and recurrent BCCs to be 1.0% and 5.6%, respectively [12,20]. Comparatively, standard excision 5-year recurrence rates were 10.1% and 17.4% for primary and recurrent BCCs, respectively [12,20]. A prospective study from the Netherlands of high-risk facial BCCs treated with MMS versus standard excision with minimum 10-years follow up revealed recurrence rates of 4.4% after MMS versus 12.2% after standard excision for primary BCCs, although this data did not reach significance (p = 0.100). For recurrent BCCs, recurrence rates were 3.9% for MMS and 13.5% for standard excision at 10 years (p = 0.023) [21]. Interestingly, 56% of primary BCC recurrences and 14% of recurrent BCC recurrences occurred after 5-years follow up [21]. More recently, a Cochrane Database Systematic Review of RCTs on BCC interventions concluded that MMS provided the lowest recurrence rates for BCC compared to standard excision and other non-surgical treatments [22]. In 2012, the American Academy of Dermatology, the American College of Mohs Surgery, the American Society for Dermatologic Surgery Association, and the American Society for Mohs Surgery developed an appropriate use criteria (AUC) for MMS based on tumor and patient characteristics [23]. These criteria stratify tumors into three groups (appropriate, uncertain, and inappropriate) based on tumor subtype, location, size, host immune status, and primary vs. recurrent tumor [23]. These clinicopathological factors influence the number of stages of Mohs surgery required to treat basal cell carcinoma. The MMS AUC is useful for clinicians in determining which BCCs are amenable to standard surgical excision vs. MMS.

2.3. Electrodesiccation and Curettage

Electrodesiccation and curettage (ED&C) is a suitable treatment for many low-risk BCCs [16]. The technique requires local anesthesia, two to three cycles of sharp curettage and electrodesiccation of the tumor to firm dermis [16]. The operator must distinguish by feel during curettage, the firm dermis from friable tumor tissue [16]. In certain anatomic areas, ED&C is criticized for operator-dependent results, higher recurrence rates, and cosmetically unappealing scars [24]. Patients with NMSC treated with ED&C have reported inferior cosmetic outcomes to surgical excision or MMS [25]. Recurrence rates range from 3% to 27% for BCCs treated by ED&C [12,13,24,26,27]. Recurrences occurred more often in tumors treated on the nose, paranasal areas, and forehead [24,28]. Aggressive BCC histologic subtypes, such as infiltrative, desmoplastic, morpheaform, or micronodular, displayed higher recurrence rates following ED&C of 27% at 6.5 years [29]. Size also predicted treatment efficacy, with clearance rates at 5-years of 84% for lesions larger than 2 cm and 98.8% for lesions smaller than 1 cm [30]. Other studies have recommended ED&C for BCCs smaller than 6-mm with high cure-rates [31]. Terminal hair-bearing sites also display higher recurrence rate following ED&C [32,33]. ED&C of BCCs should thus be considered for low-risk BCCs in non-facial, non-terminal hair-bearing, and less cosmetically sensitive areas [16]. Close follow up is recommended after ED&C to monitor the scar for tumor recurrence.

2.4. Cryotherapy

Cryotherapy is considered a suitable treatment for low-risk BCCs characterized by superficial growth pattern, small size, and low-risk anatomic locations of the trunk and extremities. In one study, the low-risk superficial BCC (sBCC) subtype demonstrated 1-year cure rates of 95.7% when treated with ED&C, compared to 100% with cryotherapy treatment [34]. A phase III clinical trial comparing photodynamic therapy to cryotherapy for treatment of BCCs showed 1 year recurrence rates of 15% for cryotherapy and 25% for PDT [35]. A randomized controlled trial on BCC treatment modalities showed 5-year recurrence rates of 8.2% with standard excision with 3-mm margins and 19.6% for curettage and two freeze-thaw cycles of cryotherapy [14].

2.5. Neodymium-Doped Yttrium Aluminum Garnet (Nd:YAG) Laser Ablation

Laser devices have been adopted recently for treating cutaneous malignancies. A pilot study using the neodymium-doped yttrium aluminum garnet (Nd:YAG) 1064 nm laser (80–120 J/cm2, 10 ms, 5-mm spot, 4-mm clinical margin, and no cooling) showed a 92% histologic cure rate at 1 month for non-facial BCCs less than 1.5 cm [36]. An expanded study of 31 tumors treated with 1064 nm Nd:YAG (125–140 J/cm2, 7–10 ms, 5-to 6-mm spot, 5-mm clinical margin, and no cooling) showed a 90% cure rate at 1-month histologic assessment [37]. Long term follow-up in these studies was limited and operation of the Nd:YAG laser required knowledge of laser endpoints and requisite settings [36,37]. A more recent study coupling Nd:YAG laser with optical coherence tomography (OCT) to treat 119 BCCs initially and again in 2 months, demonstrated clearance of all tumors [38]. The clinical recurrence rate at 1 year was 1.7% [38]. A large systematic review and meta-analysis of Nd:YAG laser treated BCCs (n = 3286) had a recurrence rate of 3.1% at median follow-up of 7.9 years [39].
Scars from Nd:YAG laser treatments were noted as superior to surgical scars and tumor clearance persisted at 9 months [40]. Other laser devices, including the pulsed dye laser (PDL), have been used for BCC treatment but with inferior results to the Nd:YAG laser [39,41]. Laser-assisted drug delivery and antibody-targeted gold nanoparticle laser-assisted BCC treatment are also being studied [41].

2.6. Carbon Dioxide Laser Ablation

Higher energy pulsed carbon dioxide (CO2) 10,600 nm lasers are another option for treating superficial, multifocal BCCs. In a small cohort of 17 BCCs undergoing pulsed CO2 laser treatment (500 mJ, 2-4W, 4-mm margin, 3 mm collimated handpiece, 3 passes), complete tumor clearance by histologic assessment was observed [42]. A retrospective review of 61 sBCCs and nBCCs treated with CO2 laser ablation revealed a 97% cure rate at a mean 3.4-years follow-up [43]. However, a systematic review and meta-analysis of CO2 laser ablation of 904 BCCs showed a 9.4% recurrence rate at median follow-up of 2.1 years [39]. An obvious caveat in comparing these studies is the variable laser settings and ablation protocols employed.
The primary shortcoming of laser ablation compared to surgical modalities is the lack of post-treatment margin assessment. For low-risk BCC treatment, CO2 laser ablation has been coupled with reflectance confocal microscopy (RCM) to guide targeted treatment and to confirm tumor ablation via cellular-level resolution [44,45]. A prospective study of 22 low-risk BCCs treated with a single pass of CO2 laser ablation showed RCM-imaged residual tumor in 22.7% cases [45]. Additional passes were administered until imaged tumor clearance was achieved, which was maintained at median follow-up of 28.5 months [45]. Pairing CO2 laser ablation with RCM is a novel treatment approach with studies underway to identify optimal ablation parameters and imaging protocols, validated by histology [44].
Fractional ablative CO2 laser can also be combined with topical medications, such as 5-fluorouracil (5-FU), to enhance percutaneous drug delivery [41]. When coupled with 5-FU, a maximum depth of drug uptake of 1.5 mm was noted an hour after treatment [41]. Carbon dioxide laser ablation followed by topical 5-FU under occlusion for a week resulted in a 71% histological clearance rate of sBCCs at 4 to 8-weeks, which is comparable to topical 5-fluorouracil monotherapy [41]. Carbon-dioxide laser-assisted drug delivery with other topical medications, such as cisplatin 0.1% solution, is being studied [41]. Carbon-dioxide laser has also been proposed in combination with photodynamic therapy, with overall good results [46].

3. Topical, Intralesional, and Field Treatments

3.1. Topical 5-Fluorouracil

5-fluorouracil (5-FU) is an antimetabolite that impairs DNA replication by blocking the synthesis of thymidylate. The 5-FU 5% cream twice daily for 3–12 weeks is FDA-approved for sBCC treatment [47]. The National Comprehensive Cancer Network (NCCN) Guidelines recommend topical 5-FU for sBCCs in low-risk locations with low-risk features [16]. Some studies have reported sBCC cure rates as high as 90% with topical 5-FU [48]. However, a large, RCT reported lower and less durable efficacy, with 5-year tumor-free survival probability of 70% (95% CI = 62.9–76.0), compared to imiquimod at 80.5% (95% CI = 74.0–85.6 [49]. For the sBCC treatment, a noninferiority randomized controlled trial showed that tumor-free survival at 3-years for 5-FU was 68.2% (95% CI = 58.1–76.3), deemed noninferior to methyl aminolevulinate photodynamic therapy (MAL-PDT) at 58.0% (95% CI = 47.8–66.9) but inferior to imiquimod at 79.7% (95% CI 71.6–85.7) [50]. Treatment with 5-FU for sBCC in low-risk anatomic locations is well-tolerated with common adverse events being erythema and erosion [16].

3.2. Topical Imiquimod

Imiquimod is an immunomodulator that antagonizes tumorigenesis in BCC by innate and adaptive immune activation through toll-like receptor (TLR) 7 and 8 agonism and proinflammatory cytokine signaling [51]. Imiquimod also downregulates GLI expression through protein kinase A (PKA) activation [52,53]. The 5% topical formulation is FDA-approved for sBCCs smaller than 2-cm in diameter on the trunk, neck, or extremities of immunocompetent adults [54].
Imiquimod treatment displays different tumor clearance rates for treating different BCC histological subtypes. A prospective trial showed a 5-year cure rate for sBCC treatment at 85% [55]. A phase III randomized controlled trial treating sBCC and nBCC showed an 84% cure rate at 3-years [56]. A systematic review described tumor clearance rates of 43 to 100% for sBCC (n = 1482 tumors), 42 to 100% for nBCC (n = 438 tumors) and 56 to 63% for infiltrative BCCs (iBCC; n = 43 tumors) [57]. The SINS trial (a multi-center, non-inferiority randomized controlled trial) established imiquimod 5% cream as less effective compared to standard excision for low-risk BCCs, with tumor clearance rates of 84% and 98% at 5-year follow up [58,59]. A recent Cochrane database systematic review of 52 RCTs with 6690 participants with nBCC or sBCC, showed that imiquimod compared to surgical excision resulted in more recurrences at 3 years (16.4% versus 1.6%) and 5 years (17.5% versus 2.3%) [22,58]. Interestingly, the rate of good/excellent cosmetic outcomes for imiquimod compared to surgical excision at 3 years was 60.6% versus 35.6% when assessed by investigators; however, no significant cosmetic differences were noted by trial participants at 6-months or 3-years post treatment [22,58]. Topical imiquimod is an acceptable treatment for low-risk BCCs, with excellent cosmetic outcomes. It is overall well-tolerated with common adverse effects being skin irritation, erythema, and rarely erosion [16].

3.3. Topical Hedgehog Inhibitors

Hedgehog pathway inhibitors (HPIs) are desirable therapeutics for their targeted molecular inhibition of aberrantly overactive Hh signaling, a major driver of BCC tumorigenesis [60]. In a double-blind, randomized, vehicle-controlled study of 8 patients with BCNS harboring 27 BCC lesions, sonidegib (LDE225) 0.75% cream compared to vehicle applied daily for 4 days resulted in a complete response in 23%, a partial response in 69%, and no response in 8% of treated BCCs [61]. Up to a 16-fold reduction in GLI1, GLI2, and PTCH2 expression was noted in most lesions in the treatment group [61]. However, clinical efficacy has been notably lower in other studies of topical HPIs. A phase II study of sporadic nBCC and sBCC treated with topical sonidegib was terminated early due to lack of efficacy [62]. A phase I study for a novel topical HPI, CUR61414, also did not show efficacy [63]. Drug formulation, penetrance, and rapid drug metabolism are potential culprits. Topical HPIs are still being researched even though they are not yet clinically available. This is because local drug delivery is expected to avoid substantial adverse effects, which frequently cause patients on systemic HPIs to discontinue treatment. Calienni et al. proposed a nano-drug delivery system for topical vismodegib, though not yet used in humans [64]. A phase II study on patidegib 2% gel daily for 12 weeks revealed greater tumor reduction compared to vehicle gel (p = 0.038) (NCT02828111) [65]. Patidegib 2% or 4% gel is being evaluated in several clinical trials for patients with BCNS and in non-syndromic patients with high frequency BCCs with results pending (NCT02762084, NCT02828111) [65,66]. A phase III study on patidegib gel 2% for BCCs in 174 BCNS patients was completed in December 2020, with results pending (NCT03703310) [67].

3.4. Histone Deacetylase Inhibitors (Vorinostat, Remetinostat)

Histone deacetylases (HDAC) enhance GLI function in the Hh pathway and are promising targets for BCC therapeutics [68]. The topical pan-histone deacetylase inhibitor (HDACi), remetinostat 1% gel applied three times daily for six weeks, was evaluated in a phase II open trial of 25 patients with 33 BCCs [69]. The overall response rate (ORR), defined as a 30% or greater decrease in tumor diameter at week 8, was 69.7% [69]. Pathologic resolution was seen in 54.8% of tumors, but response varied by histological subtype with 100% of sBCCs, 68.2% of nBCCs, and 66.7% of iBCCs responding [69]. No serious adverse events were reported. The most common side effect was treatment site dermatitis [69].
Vorinostat is another HDACi, FDA-approved for cutaneous T-cell lymphoma treatment [70]. It has been studied in advanced BCCs that circumvent SMO inhibition via GLI activation [71]. Both Vorinostat and NL-103, a synthetic chimeric compound with elements of vorinostat and vismodegib, were able to overcome SMO resistance by concurrent inhibition of HDAC function and Hedgehog signaling [71]. However, vorinostat is associated with significant adverse side effects. In a phase III trial of patients with cutaneous T-cell lymphoma, 41% of patients had grade 3/4 adverse events. In a few patients, death was attributable to treatment [72]. Systemic HDAC inhibitors have a broad side effect profile and are thus reserved for severe and refractory BCC disease only [68].

3.5. Radiation Therapy

A range of accepted radiation techniques, characteristics, and depth of therapeutic dose delivery exist. However, only therapeutic photons and electrons are endorsed by the NCCN for definitive radiation therapy (RT). Practice patterns with other modalities, such as brachytherapy and superficial X-rays, differ substantially worldwide [16]. Comparisons of heterogeneous studies on different types of RT for BCCs with different characteristics is therefore challenging, with tumor recurrence rates range widely from 3–15% at 5 years in the examined studies [13,21,73,74,75].
As primary treatment, RT is considered for local low-risk and high-risk BCCs in nonsurgical candidates and in cases when surgery would cause substantial morbidity [76]. A 10-year retrospective cohort study of BCCs treated with primary RT (comprised of electrons 19%, photons <2%, superficial X-rays 60%, or combination electrons and superficial X-rays 20%) exhibited overall tumor control rate of 92% at 2 years. Recurrent BCCs treated had a lower control rate of 86% [76]. A recent systematic review and network meta-analysis of 40 randomized trials and five nonrandomized studies comparing primary BCC treatment showed statistically equivalent tumor recurrence rates for external beam radiation (3.5%), standard excision (3.8%), and MMS (3.8%) [77]. A 10-year single center, retrospective analysis of 712 nodular BCCs (nBCC) and sBCCs treated with superficial X-ray therapy reported recurrence rates at 2 and 5 years of 2% and 4.2%, respectively [78]. Size greater than 2-cm and male sex were associated higher recurrence rates [78].
For adjuvant BCC treatment, NCCN guidelines recommend RT for significant perineural invasion or for unattainable clear margins after MMS or complete circumferential peripheral and deep margin assessment [16]. The American Society for Radiation Oncology (ASTRO) task force conducted a systematic review and recommend RT with curative intent for nonsurgical candidates and for positive margins when further surgery is not feasible or would cause cosmetic and functional morbidity [79]. Tumors with negative margins but high-risk features, such as perineural invasion or invasion of muscle, cartilage, or bone may also be treated with RT [79]. In a study of 89 patients with BCC and perineural invasion receiving adjuvant RT, 91% of patients had relapse-free survival at 5 years [80].
Radiation therapy side effects can include radiation dermatitis, depigmentation, telangiectasias, among other long term sequelae [77,81]. The risk of secondary carcinogenesis for RT treatment of BCC is not known. Patients with heightened sensitivity to radiation (i.e., homozygous ataxia telangiectasia, aplastic anemia), syndromic predisposition to carcinogenesis (i.e., Li Fraumeni, xeroderma pigmentosum, BCNS), or poorly-controlled connective tissue disease (i.e., scleroderma) are generally precluded from RT [79].
With regard to cosmetic outcomes, a meta-analysis of 58 studies and 21,000 patients reported both brachytherapy and MMS as having improved cosmesis over external beam radiation and standard excision; recurrence rates at one year were similar across the modalities [82]. Another RCT of facial BCCs treated surgically or with RT revealed superior cosmetic results as assessed by dermatologists and patients at 4-years post-treatment [83].

3.6. Photodynamic Therapy

Photodynamic therapy (PDT) is a selective anti-cancer treatment in which an administered photosensitizer is preferentially taken up by neoplastic cells [84]. Targeted irradiation with near infrared light results in reactive oxygen species production causing cellular apoptosis [84]. PDT using 5-aminolevulinic acid (ALA) and methyl aminolevulinic acid (mALA), which are prodrugs of protoporphyrin IX, is an off-label treatment for sBCC [84]. Although mALA is no longer produced in the US, mALA, and ALA are approved for sBCC and thin nBCC in Europe [84].
ALA and mALA boast similar efficacy in treating sBCC and thin nBCC, less than 2-cm in diameter and less than 2-mm in histologic thickness [85]. Cure rates range from 60–100% for nBCC and sBCC treatment, with higher complete response rates in sBCC, truncal location, absent ulceration, and thickness less than 0.5-mm [86]. A phase III study as well as a systematic review and meta-analysis found PDT superior in cosmetic outcomes, but inferior in disease control compared to standard excision and cryotherapy [35,87]. One meta-analysis of 23 studies found PDT comparable to imiquimod for sBCC treatment efficacy at 1 year follow up [88]. However, a RCT of 601 patients with sBCC reported tumor-free survival at 5-years to be 62.7% with PDT and 80.5% for imiquimod [49]. Despite the lower efficacy compared to other modalities, patients with numerous BCCs or a predisposing syndrome such as BCNS may benefit from PDT field treatments [89].
Currently, PDT typically employs near infrared wavelengths for optimal penetration depth. Intralesional PDT to enhance delivery depth has also been described for treating different histological subtypes of BCCs although efficacy was comparable to conventional modalities [90]. PDT still remains an alternative treatment in nonsurgical candidates or for low-risk BCCs in which cosmesis is a priority [16]. Common adverse reactions such as pain, pruritus, erythema, and edema are usually limited to the treated area [16].

4. Systemic Therapies

4.1. Vismodegib

Vismodegib (Erivedge Capsule, Genentech Inc., San Francisco, CA, USA) is a small molecule inhibitor of SMO targeting Hh pathway over-activation present in most BCCs [91,92]. In the 2009 Phase I study of patients with laBCC or mBCC on vismodegib, the objective response rate (ORR) was 58% with median duration of response (DOR) of 12.8 months [93]. Following this, pharmacokinetic studies determined that vismodegib 150 mg daily produced the highest plasma concentrations [94]. In 2012, Vismodegib 150 mg daily was FDA-approved for treating laBCC and mBCC following robust evidence of efficacy in the Erivance clinical trials (a multicenter, international, single arm, two-cohort, non-randomized study) which demonstrated an ORR of 30% in patients with mBCC and 43% with laBCC [91]. Within the laBCC cohort, 21% demonstrated a complete response (CR). Median duration of response was 7.6 months in both cohorts. Adverse events (AEs) occurring in more than 30% of patients included muscle cramps (68%), alopecia (63%), and dysgeusia (51%). Nearly all patients experienced at least one adverse event, the majority being grade 2 or lower [91]. At 21-month follow-up, respective ORR was 48% (median DOR 9.5 months) and 33% (median DOR 7.6 months) in the laBCC and mBCC groups [95]. Independent analysis of the Erivance data revealed that 65% of laBCC demonstrated significant improvement, while 11% of the cohort experienced progression on vismodegib [96]. An evaluation of vismodegib prophylaxis in BCNS patients (a double-blind, randomized, phase II study) showed decreased BCC incidence and size compared to placebo [97]. Despite the improvement noted with vismodegib in highly morbid BCC disease, 20% of mBCC patients in the Erivance trial progressed, which underscores drug resistance mechanisms that synergistic treatment regimens may address [91]. Vismodegib as neoadjuvant therapy prior to surgery has also been used. An open-label, single arm trial of large (average 12.6 cm2 area) and high-risk, operable BCCs showed a 27% decreased surgical defect area after preoperative vismodegib treatment for 3-6 months [98]. Vismodegib is effective for advanced BCCs, although treatment resistance to HPIs can be seen. Vismodegib coupled with other therapeutics are currently being studied.

4.2. Sonidegib

Another small molecule HPI, sonidegib (Odomzo, Novartis Int. AG, Basel, Switzerland) was approved in 2015 for treating laBCC [99]. Sonidegib was identified in 2010, noted for high tissue penetration and oral bioavailability, blood-brain barrier penetration, and potent anti-tumor activity in medulloblastoma allograft models [100,101]. Phase I studies in patients with medulloblastoma or advanced BCC identified the maximal tolerable dose to be 800 mg daily and 250 mg twice daily. The AEs were comparable to vismodegib AEs, except for reversible grade 3/4 Creatine Kinase (CK) elevation noted in 19% of patients when doses exceeded the maximal tolerable dose [102]. Primary efficacy analysis of the BOLT trial, a randomized, double-blind, multicenter, phase II study, evaluated a 800 mg and 200 mg dose of sonidegib, which showed equivalent ORRs of 34% and 36% respectively, in laBCC and mBCC groups at median follow up of 13.9 months [103]. At 42-month follow-up, the 200 mg dose had an ORR of 56.1% in laBCC and 7.7% in mBCC with a respective DOR of 26.1 and 24 months [104]. Sonidegib is an effective treatment for laBCC, although mutations conferring resistance are seen in HPI-treated advanced BCCs. In a small trial of patients with vismodegib-resistant advanced BCCs treated with sonidegib for a median of 6-weeks, over half of the patients had progressive disease [105].

4.3. Cemiplimab

Cemiplimab, a fully human monoclonal antibody against the PD1 receptor, is an FDA-approved immune checkpoint inhibitor for treating locally advanced cutaneous squamous cell carcinoma (laSCC) and metastatic cutaneous squamous cell carcinoma (mSCC) not amenable to surgery or radiation therapy [106]. In February 2021, cemiplimab garnered FDA approval for treating locally-advanced and metastatic BCC, thereby providing HPI-refractory patients a second-line treatment option [106,107]. BCCs harbor a high mutational burden and are thus expected to respond well to broad anti-tumor immune activity, elicited by immune checkpoint inhibitors such as cemiplimab. In an open label, multicenter, single arm clinical trial of patients with laBCC and mBCC, ORR was noted in 31% of patients by independent review [106]. Complete response was seen in 6% of patients. Forty-eight percent of patients had grade 3–4 adverse side effects, most commonly hypertension. Thirty-five percent of patients exhibited severe adverse side effects. There were no treatment-related deaths. Despite the side effect profile, cemiplimab demonstrated clinical efficacy as a second-line treatment for advanced BCC, whether locally-advanced or metastatic, particularly in those individuals that demonstrate progression, stable disease, or intolerable side effects on HPIs [106,108].

5. Investigational Drugs

5.1. Taladegib (LY2940680)

Taladegib (LY2940680) is a competitive SMO antagonist that inhibits the activity of vismodegib-resistant SMO mutants at D473H [109]. In a phase I study, taladegib treating laBCC and mBCC, whether treatment-naïve or refractory to HPIs, demonstrated efficacy with 46.8% of patients exhibiting a complete or partial response at 10.2 months duration of response [110]. Like other HPIs, common adverse events included dysgeusia, fatigue, nausea, and muscle spasms [110].

5.2. Patidegib (TAK-441)

Patidegib is a semi-synthetic, topical derivative of cyclopamine and functions by SMO inhibition. A phase I trial showed that patidegib 160 mg daily was well-tolerated [111]. A multi-center, double-blind, randomized, vehicle-controlled phase 2 study demonstrated clinical efficacy and safety of patidegib topical 2% and 4% gel for stage I BCCs in patients with BCNS (NCT02762084) [65]. In a subsequent study, patidegib 4% gel showed higher clinical and molecular response [66]. An additional phase 2 study on patidegib 2% gel for BCC prevention in patients with non-syndromic, high frequency BCCs, is pending results (NCT04155190) [66]. A completed multicenter, randomized, double-blind, vehicle-controlled, Phase 3 study evaluating patidegib 2% gel twice daily for over 1 year for reducing disease burden in 174 BCNS patients is pending results (NCT03703310) [67]. Another open-label extension study is pending results on treatment-related adverse events associated with twice daily application in 107 participants (NCT04308395) [112].

5.3. LEQ506

LEQ506 is a potent second-generation SMO antagonist with a pyridazine core evaluated in clinical trials from 2010 to 2015 (NCT01106508) [113,114]. In medulloblastoma allograft models, LEQ506 mediated near complete and sustained GLI1 mRNA inhibition [114]. In depilated murine models it decreased GLI1 expression by 80–90% and PTCH1 mRNA expression by 60–70% after topical application [115]. Phase I studies showed safety of LEQ506 daily for three weeks in adults with advanced solid tumor, recurrent or refractory medulloblastoma, laBCC or mBCC [113]. The maximal tolerated dose was 400 mg with limiting toxicities including fatigue, dyspnea, and grade 3/4 elevations in CK, AST, ALT, and uric acid [113].

5.4. ZSP 1602

ZSP 1602 is an oral second-generation SMO inhibitor evaluated in phase I clinical trials from 2019 to 2021 (NCT03734913) [116]. Part 1 included participants with advanced solid tumors including BCC and medulloblastoma, regardless of SMO or GLI1 expression levels. Part 2 Cohort A included participants with esophagogastric adenocarcinoma with SMO or GLI1 overexpression. Part 2 Cohort B included participants with BCC, small cell lung cancer, neuroendocrine neoplasms, and glioblastoma with SMO or GLI1 overexpression. Results from the trial are pending [116].

5.5. CK2 Inhibitors (CX-4945)

Casein kinase 2 (CK-2) is a serine/threonine kinase in Hh pathway activation. It prevents GLI transcription factor degradation and phosphorylates SMO thereby enhancing its activity and cell surface concentration [117,118]. CK-2 inhibition has been shown to reduce GLI1 expression and Hh pathway overactivation in many tumors, including lung cancer, lung cancer cell lines, and medulloblastoma [119,120,121]. A phase I open label trial studying CK-2 inhibitor (CX-4945) to treat SMO inhibitor-resistant laBCC and mBCC is underway (NCT03897036) [122]. CX-4945 inhibition is sufficiently downstream in the Hh pathway, which reduces the likelihood of off-target effects and acquired resistance through downstream mutations.

5.6. Itraconazole

Itraconazole is an antifungal medication that exhibits SMO antagonism [123]. Topical and oral itraconazole have decreased BCC tumor size in mouse models [124]. In an open-label, phase II trial of oral itraconazole for BCC treatment, 19 of 29 patients were treated with oral itraconazole 200 mg twice daily for 1 month, or 100 mg twice daily for 2.3 months. Itraconazole markedly decreased cell proliferation by 45% (measure by Ki67 tumor proliferation), Hh pathway activity by 65% (measured by GLI1 mRNA expression), and tumor area by 24% [125]. Four of 8 patients with multiple BCCs achieved partial response and the other 4 had stable disease [125]. A Phase 2, open-label, placebo-controlled trial of topical itraconazole 0.7% gel applied for 1–3 months in BCC treatment did not yield reduction of GLI1 mRNA expression or tumor size [126]. Itraconazole is being studied in combination with other SMO antagonists, specifically vismodegib, sonidegib, and with arsenic trioxide [127].

5.7. GLI Antagonists (GANTS)

The terminal step of the Hh pathway results in GLI transcription factor activation, nuclear translocation, and gene expression to govern cell proliferation and the epithelial to mesenchymal transition, a critical regulatory first step in the metastatic process [128,129,130]. GLI antagonists-58 and -61 (GANTS) have been studied in cancer cell lines, and GANT-61 has demonstrated anti-tumor effects on xenograft models of neuroblastoma [131,132,133]. No clinical trials are underway for GANTS in BCC treatment.

5.8. Anti VEGFR (AIV001)

Vascular endothelial growth factor (VEGF) stimulates the angiogenesis needed for tumor growth, invasion, and metastasis [134]. VEGF expression under certain conditions is regulated by the Hh pathway [135]. Recent studies also demonstrated direct oncogenic effects of VEGF, which alters keratinocyte survival and proliferation to favor NMSC development [136]. Phase I and II open label, non-randomized trials on AIV001, a prolonged-release multi-kinase inhibitor with anti-angiogenic and antineoplastic properties for treating sBCC and nBCC (NCT04470726) [137].

5.9. Anti COX-2 TGFB SiRNA

Transforming growth factor-β (TGF-β) is a group of proteins that mediate cell proliferation, apoptosis, migration and epithelial-to-mesenchymal transition, all of which are critical in cancer progression [138]. In iBCCs, TGF-β has been identified in the peritumoral stroma and has been shown to induce peritumoral fibronectin, which promotes cell migration and adhesion needed for tumor infiltration [139]. Overexpression of cyclooxigenase-2 (COX-2), which facilitates prostaglandin formation from arachidonic acid, predicts increased BCC recurrence risk, angiogenesis, and invasive depth [140,141]. STP705, a silencing RNA (siRNA) that knocks down local expression TGF-β and COX-2, is an intralesional medication in current phase II dose-escalation open-label trials for BCC treatment (NCT04669808) [142].

5.10. Anti LAG3 Ab

Lymphocyte activation gene 3 (LAG3), also known as CD223, is an immune checkpoint receptor highly expressed on activated regulatory CD4 and CD8 T-cells, tumor infiltrating lymphocytes, B-cells, natural killer cells, and dendritic cells [143,144]. LAG3 amplifies the immunosuppressive function of T-regulatory cells [144]. LAG3 interacts predominantly with MHC-II, preventing T-cell receptor binding and precluding T-cell activation [143]. PD1 and LAG3 are commonly co-expressed on anergic peripheral T-cells, and simultaneous blockade can reverse T-cell anergy and enhance anti-tumor activity [144,145]. This effect was seen in a randomized, double-blind, phase II-III study assessing clinical efficacy of combined treatment with relatlimab, a LAG-3-blocking antibody, and nivolumab, a PD-1-blocking antibody versus nivolumab monotherapy in advanced melanoma patients [146]. Median progression-free survival (PFS) was significantly longer with relatlimab-nivolumab (10.1 months [95% CI 6.4 to 15.7]) compared to nivolumab monotherapy (4.6 months [95% CI 3.4 to 5.6]) [146]. Greater grade 3 or 4 treatment-related adverse events, occurring in 18.9% of patients in the relatlimab-nivolumab group were observed compared to in 9.7% of patients in the nivolumab monotherapy group [146]. The relatlimab and nivolumab combination compared to nivolumab monotherapy is currently in phase II trials for laBCC and mBCC that has progressed on anti PD(L)-1 monotherapy (NCT03521830) [147].

5.11. Intralesional Talimogene Laherparepvec (T-VEC)

Intralesional talimogene laherparepvec (T-VEC) is a genetically-modified herpes simplex virus oncolytic virus therapy directly injected into accessible tumors to enhance the local and systemic anti-tumor immune response. Selective viral replication triggers tumor cell lysis and antigen release, which activates tumor-specific effector T-cells [148]. T-VEC has been used to treat melanoma, primary cutaneous B-cell lymphoma (pCBCL), and Merkel cell carcinoma [149]. T-VEC knock-in of granulocyte macrophage colony stimulating factor (GM-CSF) can enhance local and systemic antitumor activity by stimulating other arms of the immune system [150]. A phase I, open label, single arm, single center study on intralesional T-VEC for locally-advanced SCCs, BCCs, Merkel cell carcinomas, and CTCL is ongoing(NCT03458117) [151]. A phase II trial on T-VEC with nivolumab for patients with refractory NMSC, including BCCs, is also ongoing (NCT02978625) [152].

5.12. IL2/TNFa

Intralesional therapies offer direct drug delivery to the tumor and decreased systemic drug exposure and related toxicities [153,154]. Bifikafusp alfa + onfekafusp alfa, (Daromun) consists of recombinant interleukin-2 (IL-2) and tumor necrosis factor alpha (TNF-α) fused to an L19 monoclonal antibody (L19IL2/L19TNF) [155]. IL-2 is critical for T-cell development and function, while TNF-α exhibits cytotoxicity to cancer cells [156,157]. The immunocytokine combination is in a phase II clinical study (NCT04362722) for treating high-risk, laBCC or cSCC [158]. In non-surgical candidates with stage III or IV melanoma, the drug has shown CR in 32 melanoma lesions (28.3%; 21 target and 11 non-target lesions) at week 12 [159]. A CR was seen in 7 of 13 (53.8%) non-injected lesions (4 cutaneous, 3 lymph nodes), which suggests that intralesional treatment elicits a systemic immune response [159]. Intralesional L19IL2/L19TNF had primarily low grade 1 and 2 adverse events such as injection site reactions (72.7% of patients) or low-grade fever (59% of patients) [159]. Based on these results, patients with BCC are hypothesized to respond to intralesional L19IL2/L19TNF therapy.

5.13. IFN Gamma Adenovirus

Other intratumoral oncolytic virus therapeutics include ASN-002, a replication-defective adenovirus vector that expresses recombinant human IFN-gamma gene in target cells. This decreases cell proliferation via cell cycle inhibition, apoptosis, suppression of angiogenesis, and indirect immune-mediated anti-tumor effects [160,161,162,163]. Targeted cells express IFN-gamma, thereby circumventing toxicities associated with systemic IFN-gamma therapy. An ongoing phase II open-label trial is examining intralesional ASN-002 with vismodegib for sporadic or BCNS-related BCCs (NCT04416516) [164].

6. Discussion

Treatment options for BCC range from surgical, intralesional, topical, field treatments, targeted molecular therapies, to immunotherapy. Therapeutic choice relies on comprehensive evaluation of tumor-specific risk factors, including histologic subtype, size, and anatomic location, with patient factors, such immunosuppression, surgical candidacy, therapy-related morbidity, and patient preference. For low-risk and small BCCs, several different therapeutic approaches may offer comparable results but different side effect profiles, thus a patient’s informed preference, concerns about cosmesis, invasiveness of the treatment approach, and risk of adverse events should influence collaborative discussion of treatment options. For larger BCCs, high-risk histology, high-risk anatomic location, or when surgery would result in high morbidity, systemic and targeted molecular therapies should be considered in concert with multidisciplinary input. Immunotherapeutics and targeted molecular therapies have been coupled with definitive surgical modalities, with improved outcomes. The hedgehog pathway has been identified as a major molecular pathway driving the progression of many BCCs, and advances in systemic and local therapeutics targeting this pathway have broadened treatment options for many patients with highly morbid BCC disease. Promising investigational drugs are emerging that target the diverse molecular drivers behind BCC pathogenesis. Additionally, cemiplimab immunotherapy has emerged as a promising treatment for patients who have failed hedgehog inhibitors and other targeted therapeutics. Together, these new drugs promise to broaden further the treatment options for patients with BCC and permit a more individualized approach to BCC treatment.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers14153720/s1, Table S1. Risk-stratified clinicopathologic features of basal cell carcinoma.

Author Contributions

All authors participated in the literature review, writing, editing, and review of this manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

We thank Christopher R. Deig for his constructive review of the radiation therapy section of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rogers, H.W.; Weinstock, M.A.; Feldman, S.R.; Coldiron, B.M. Incidence Estimate of Nonmelanoma Skin Cancer (Keratinocyte Carcinomas) in the US Population, 2012. JAMA Dermatol. 2015, 151, 1081–1086. [Google Scholar] [CrossRef] [PubMed]
  2. Ruiz, E.S.; Morgan, F.C.; Zigler, C.M.; Besaw, R.J.; Schmults, C.D. Analysis of national skin cancer expenditures in the United States Medicare population, 2013. J. Am. Acad. Dermatol. 2019, 80, 275–278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Roewert-Huber, J.; Lange-Asschenfeldt, B.; Stockfleth, E.; Kerl, H. Epidemiology and aetiology of basal cell carcinoma. Br. J. Dermatol. 2007, 157, 47–51. [Google Scholar] [CrossRef]
  4. Migden, M.R.; Chang, A.L.S.; Dirix, L.; Stratigos, A.J.; Lear, J.T. Emerging trends in the treatment of advanced basal cell carcinoma. Cancer Treat. Rev. 2018, 64, 1–10. [Google Scholar] [CrossRef] [PubMed]
  5. Madan, V.; Lear, J.T.; Szeimies, R.M. Non-melanoma skin cancer. Lancet 2010, 375, 673–685. [Google Scholar] [CrossRef] [Green Version]
  6. Bacchetti, T.; Salvolini, E.; Pompei, V.; Campagna, R.; Molinelli, E.; Brisigotti, V.; Togni, L.; Lucarini, G.; Sartini, D.; Campanati, A.; et al. Paraoxonase-2: A potential biomarker for skin cancer aggressiveness. Eur. J. Clin. Investig. 2020, 51, e13452. [Google Scholar] [CrossRef] [PubMed]
  7. Sharpe, H.J.; Pau, G.; Dijkgraaf, G.J.; Basset-Seguin, N.; Modrusan, Z.; Januario, T.; Tsui, V.; Durham, A.B.; Dlugosz, A.A.; Haverty, P.M.; et al. Genomic Analysis of Smoothened Inhibitor Resistance in Basal Cell Carcinoma. Cancer Cell 2015, 27, 327–341. [Google Scholar] [CrossRef] [Green Version]
  8. Bonilla, X.; Parmentier, L.; King, B.; Bezrukov, F.; Kaya, G.; Zoete, V.; Seplyarskiy, V.B.; Sharpe, H.J.; McKee, T.; Letourneau, A.; et al. Genomic analysis identifies new drivers and progression pathways in skin basal cell carcinoma. Nat. Genet. 2016, 48, 398–406. [Google Scholar] [CrossRef]
  9. Gualdi, G.; Monari, P.; Apalla, Z.; Lallas, A. Surgical treatment of basal cell carcinoma and squamous cell carcinoma. G. Ital. Dermatol. Venereol. 2015, 150, 435–447. [Google Scholar]
  10. Wolf, D.J.; Zitelli, J.A. Surgical margins for basal cell carcinoma. Arch. Dermatol. 1987, 123, 340–344. [Google Scholar] [CrossRef]
  11. Rhodes, L.E.; De Rie, M.A.; Leifsdottir, R.; Yu, R.C.; Bachmann, I.; Goulden, V.; Wong, G.A.E.; Richard, M.-A.; Anstey, A.; Wolf, P. Five-Year Follow-up of a Randomized, Prospective Trial of Topical Methyl Aminolevulinate Photodynamic Therapy vs Surgery for Nodular Basal Cell Carcinoma. Arch. Dermatol. 2007, 143, 1131–1136. [Google Scholar] [CrossRef] [PubMed]
  12. Rowe, D.E.; Carroll, R.J.; Day, C.L.J. Long-Term Recurrence Rates in Previously Untreated (Primary) Basal Cell Carcinoma: Implications for Patient Follow-Up. J. Dermatol. Surg. Oncol. 1989, 15, 315–328. [Google Scholar] [CrossRef] [PubMed]
  13. Thissen, M.R.T.M.; Neumann, M.H.A.; Schouten, L. A Systematic Review of Treatment Modalities for Primary Basal Cell Carcinomas. Arch. Dermatol. 1999, 135, 1177–1183. [Google Scholar] [CrossRef] [PubMed]
  14. Kuijpers, D.I.M.; Thissen, M.R.T.M.; Berretty, P.J.M.; Ideler, F.H.L.B.; Nelemans, P.J.; Neumann, M.H.A.M. Surgical Excision versus Curettage plus Cryosurgery in the Treatment of Basal Cell Carcinoma. Dermatol. Surg. 2007, 33, 579–587. [Google Scholar] [CrossRef] [PubMed]
  15. 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] [PubMed]
  16. Bichakjian, C.K.; Olencki, T.; Aasi, S.Z.; Alam, M.; Andersen, J.S.; Berg, D.; Bowen, G.M.; Cheney, R.T.; Daniels, G.A.; Glass, L.F.; et al. Basal Cell Skin Cancer, Version 1.2016, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2016, 14, 574–597. [Google Scholar] [CrossRef] [PubMed]
  17. Nisticò, S.P.; Bennardo, L.; Sannino, M.; Negosanti, F.; Tamburi, F.; Del Duca, E.; Giudice, A.; Cannarozzo, G. Combined CO 2 and dye laser technique in the treatment of outcomes due to flap necrosis after surgery for basal cell carcinoma on the nose. Lasers Surg. Med. 2021, 54, 523–529. [Google Scholar] [CrossRef] [PubMed]
  18. Campagna, R.; Pozzi, V.; Sartini, D.; Salvolini, E.; Brisigotti, V.; Molinelli, E.; Campanati, A.; Offidani, A.; Emanuelli, M. Beyond Nicotinamide Metabolism: Potential Role of Nicotinamide N-Methyltransferase as a Biomarker in Skin Cancers. Cancers 2021, 13, 4943. [Google Scholar] [CrossRef]
  19. Lawrence, C.M. Mohs’ micrographic surgery for basal cell carcinoma. Clin. Exp. Dermatol. 1999, 24, 130–133. [Google Scholar] [CrossRef]
  20. Rowe, D.E.; Carroll, R.J.; Day, C.L. 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]
  21. van Loo, E.; Mosterd, K.; Krekels, G.A.; Roozeboom, M.H.; Ostertag, J.U.; Dirksen, C.D.; Steijlen, P.M.; Neumann, H.M.; Nelemans, P.J.; Kelleners-Smeets, N.W. Surgical excision versus Mohs’ micrographic surgery for basal cell carcinoma of the face: A randomised clinical trial with 10year follow-up. Eur. J. Cancer 2014, 50, 3011–3020. [Google Scholar] [CrossRef] [PubMed]
  22. Thomson, J.; Hogan, S.; Leonardi-Bee, J.; Williams, H.C.; Bath-Hextall, F.J. Interventions for basal cell carcinoma of the skin. Cochrane Database Syst. Rev. 2020, 2020, CD003412. [Google Scholar] [CrossRef]
  23. Connolly, S.M.; Baker, D.R.; Coldiron, B.M.; Fazio, M.J.; Storrs, P.A.; Vidimos, A.T.; Zalla, M.J.; Brewer, J.D.; Begolka, W.S.; Berger, T.G.; et al. AAD/ACMS/ASDSA/ASMS 2012 appropriate use criteria for Mohs micrographic surgery: A report of the American Academy of Dermatology, American College of Mohs Surgery, American Society for Dermatologic Surgery Association, and the American Society for Mohs Surgery. J. Am. Acad. Dermatol. 2012, 67, 531–550. [Google Scholar] [CrossRef] [PubMed]
  24. Kopf, A.W.; Bart, R.S.; Schrager, D.; Lazar, M.; Popkin, G.L. Curettage-electrodesiccation treatment of basal cell carcinomas. Arch. Dermatol. 1977, 113, 439–443. [Google Scholar] [CrossRef] [PubMed]
  25. Galles, E.; Parvataneni, R.; Stuart, S.E.; Linos, E.; Grewal, S.; Chren, M.-M. Patient-reported outcomes of electrodessication and curettage for treatment of nonmelanoma skin cancer. J. Am. Acad. Dermatol. 2014, 71, 1026–1028. [Google Scholar] [CrossRef] [Green Version]
  26. Barlow, J.O.; Zalla, M.J.; Kyle, A.; DiCaudo, D.J.; Lim, K.K.; Yiannias, J.A. Treatment of basal cell carcinoma with curettage alone. J. Am. Acad. Dermatol. 2006, 54, 1039–1045. [Google Scholar] [CrossRef]
  27. Julian, C.; Bowers, P.; Pritchard, C. A comparative study of the effects of disposable and Volkmann spoon curettes in the treatment of basal cell carcinoma. Br. J. Dermatol. 2009, 161, 1407–1409. [Google Scholar] [CrossRef]
  28. Rodriguez-Vigil, T.; Vázquez-López, F.; Perez-Oliva, N. Recurrence rates of primary basal cell carcinoma in facial risk areas treated with curettage and electrodesiccation. J. Am. Acad. Dermatol. 2007, 56, 91–95. [Google Scholar] [CrossRef]
  29. Blixt, E.; Nelsen, D.; Stratman, E. Recurrence Rates of Aggressive Histologic Types of Basal Cell Carcinoma After Treatment with Electrodesiccation and Curettage Alone. Dermatol. Surg. 2013, 39, 719–725. [Google Scholar] [CrossRef]
  30. Spiller, W.F.; Spiller, R.F. Treatment of basal cell epithelioma by curettage and electrodesiccation. J. Am. Acad. Dermatol. 1984, 11, 808–814. [Google Scholar] [CrossRef]
  31. Silverman, M.K.; Kopf, A.W.; Grin, C.M.; Bart, R.S.; Levenstein, M.J. Recurrence Rates of Treated Basal Cell Carcinomas: Part 2: Curettage-Electrodesiccation. J. Dermatol. Surg. Oncol. 1991, 17, 720–726. [Google Scholar] [CrossRef] [PubMed]
  32. Goldman, G. The current status of curettage and electrodesiccation. Dermatol. Clin. 2002, 20, 569–578. [Google Scholar] [CrossRef]
  33. Lewin, J.M.; Carucci, J. Advances in the management of basal cell carcinoma. F1000Prime Rep. 2015, 7, 53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Backman, E.; Polesie, S.; Berglund, S.; Gillstedt, M.; Sjöholm, A.; Modin, M.; Paoli, J. Curettage vs. cryosurgery for superficial basal cell carcinoma: A prospective, randomised and controlled trial. J. Eur. Acad. Dermatol. Venereol. 2022. [Google Scholar] [CrossRef]
  35. Wang, I.; Bendsoe, N.; Klinteberg, C.A.; Enejder, A.M.; Andersson-Engels, S.; Svanberg, S.; Svanberg, K. Photodynamic therapy vs. cryosurgery of basal cell carcinomas: Results of a phase III clinical trial. Br. J. Dermatol. 2001, 144, 832–840. [Google Scholar] [CrossRef]
  36. Ortiz, A.E.; Anderson, R.R.; Avram, M.M. 1064 nm long-pulsed Nd:YAG laser treatment of basal cell carcinoma. Lasers Surg. Med. 2015, 47, 106–110. [Google Scholar] [CrossRef] [PubMed]
  37. Ortiz, A.E.; Anderson, R.R.; DiGiorgio, C.; Jiang, S.I.B.; Shafiq, F.; Avram, M.M. An expanded study of long-pulsed 1064 nm Nd:YAG laser treatment of basal cell carcinoma. Lasers Surg. Med. 2018, 50, 727–731. [Google Scholar] [CrossRef] [PubMed]
  38. Markowitz, O.; Bressler, M.Y. Combining Nd:YAG laser with optical coherence tomography for nonsurgical treatment of basal cell carcinoma. Lasers Surg. Med. 2021, 54, 105–112. [Google Scholar] [CrossRef] [PubMed]
  39. Sharon, E.; Snast, I.; Lapidoth, M.; Kaftory, R.; Mimouni, D.; Hodak, E.; Levi, A. Laser Treatment for Non-Melanoma Skin Cancer: A Systematic Review and Meta-Analysis. Am. J. Clin. Dermatol. 2020, 22, 25–38. [Google Scholar] [CrossRef] [PubMed]
  40. Ahluwalia, J.; Avram, M.M.; Ortiz, A.E. Outcomes of long-pulsed 1064 nm Nd:YAG laser treatment of basal cell carcinoma: A retrospective review. Lasers Surg. Med. 2018, 51, 34–39. [Google Scholar] [CrossRef] [Green Version]
  41. 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]
  42. Humphreys, T.R.; Malhotra, R.; Scharf, M.J.; Marcus, S.M.; Starkus, L.; Calegari, K. Treatment of Superficial Basal Cell Carcinoma and Squamous Cell Carcinoma In Situ With a High-Energy Pulsed Carbon Dioxide Laser. Arch. Dermatol. 1998, 134, 1247–1252. [Google Scholar] [CrossRef] [Green Version]
  43. Iyer, S.; Bowes, L.; Kricorian, G.; Friedli, A.; Fitzpatrick, R.E. Treatment of Basal Cell Carcinoma with the Pulsed Carbon Dioxide Laser: A Retrospective Analysis. Dermatol. Surg. 2004, 30, 1214–1218. [Google Scholar] [CrossRef] [PubMed]
  44. Hibler, B.; Sierra, H.; Cordova, M.; Phillips, W.; Rajadhyaksha, M.; Nehal, K.; Rossi, A. Carbon dioxide laser ablation of basal cell carcinoma with visual guidance by reflectance confocal microscopy: A proof-of-principle pilot study. Br. J. Dermatol. 2016, 174, 1359–1364. [Google Scholar] [CrossRef] [Green Version]
  45. Navarrete-Dechent, C.; Cordova, M.; Liopyris, K.; Yélamos, O.; Aleissa, S.; Hibler, B.; Sierra, H.; Sahu, A.; Blank, N.; Rajadhyaksha, M.; et al. Reflectance confocal microscopy-guided carbon dioxide laser ablation of low-risk basal cell carcinomas: A prospective study. J. Am. Acad. Dermatol. 2019, 81, 984–988. [Google Scholar] [CrossRef] [PubMed]
  46. Mercuri, S.R.; Brianti, P.; Dattola, A.; Bennardo, L.; Silvestri, M.; Schipani, G.; Nisticò, S.P. CO2 laser and photodynamic therapy: Study of efficacy in periocular BCC. Dermatol. Ther. 2018, 31, e12616. [Google Scholar] [CrossRef] [PubMed]
  47. Bausch Health. Efudex (5-fluorouracil) [Package Insert]. U.S. Food and Drug Administration, 14 October 2021. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/016831Orig1s063lbl.pdf (accessed on 30 April 2022).
  48. 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] [PubMed]
  49. Jansen, M.H.E.; Mosterd, K.; Arits, A.H.; Roozeboom, M.H.; Sommer, A.; Essers, B.A.; van Pelt, H.P.; Quaedvlieg, P.J.; Steijlen, P.M.; Nelemans, P.J.; et al. Five-Year Results of a Randomized Controlled Trial Comparing Effectiveness of Photodynamic Therapy, Topical Imiquimod, and Topical 5-Fluorouracil in Patients with Superficial Basal Cell Carcinoma. J. Investig. Dermatol. 2018, 138, 527–533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Roozeboom, M.H.; Arits, A.H.M.M.; Mosterd, K.; Sommer, A.; Essers, B.A.B.; De Rooij, M.J.M.; Quaedvlieg, P.J.F.; Steijlen, P.M.; Nelemans, P.J.; Kelleners-Smeets, N.W.J. Three-Year Follow-Up Results of Photodynamic Therapy vs. Imiquimod vs. Fluorouracil for Treatment of Superficial Basal Cell Carcinoma: A Single-Blind, Noninferiority, Randomized Controlled Trial. J. Investig. Dermatol. 2016, 136, 1568–1574. [Google Scholar] [CrossRef] [Green Version]
  51. Desai, T.; Chen, C.L.; Desai, A.; Kirby, W. Basic Pharmacology of Topical Imiquimod, 5-Fluorouracil, and Diclofenac for the Dermatologic Surgeon. Dermatol. Surg. 2012, 38, 97–103. [Google Scholar] [CrossRef]
  52. Bubna, A. Imiquimod—Its role in the treatment of cutaneous malignancies. Indian J. Pharmacol. 2015, 47, 354–359. [Google Scholar] [CrossRef] [Green Version]
  53. Gruber, W.; Frischauf, A.-M.; Aberger, F. An old friend with new skills: Imiquimod as novel inhibitor of Hedgehog signaling in basal cell carcinoma. Oncoscience 2014, 1, 567–573. [Google Scholar] [CrossRef] [PubMed]
  54. US Food and Drug Administration. Aldara® (Imiquimod) Cream, 5%. Silver Springs: US Food and Drug Administration, 2010. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2010/020723s022lbl.pdf (accessed on 4 April 2021).
  55. Quirk, C.; Gebauer, K.; De’Ambrosis, B.; Slade, H.B.; Meng, T.-C. Sustained clearance of superficial basal cell carcinomas treated with imiquimod cream 5%: Results of a prospective 5-year study. Cutis 2010, 85, 318–324. [Google Scholar] [PubMed]
  56. Arits, A.H.; Mosterd, K.; AB Essers, B.; Spoorenberg, E.; Sommer, A.; De Rooij, M.J.; van Pelt, H.P.; Quaedvlieg, P.J.; Krekels, G.A.; van Neer, P.A.; et al. Photodynamic therapy versus topical imiquimod versus topical fluorouracil for treatment of superficial basal-cell carcinoma: A single blind, non-inferiority, randomised controlled trial. Lancet Oncol. 2013, 14, 647–654. [Google Scholar] [CrossRef]
  57. Love, W.E.; Bernhard, J.D.; Bordeaux, J.S. Topical Imiquimod or Fluorouracil Therapy for Basal and Squamous Cell Carcinoma: A systematic review. Arch. Dermatol. 2009, 145, 1431–1438. [Google Scholar] [CrossRef]
  58. Bath-Hextall, F.; Bong, J.; Perkins, W.; Williams, H. Interventions for basal cell carcinoma of the skin: Systematic review. BMJ 2004, 329, 705. [Google Scholar] [CrossRef] [Green Version]
  59. Williams, H.C.; Bath-Hextall, F.; Ozolins, M.; Armstrong, S.J.; Colver, G.B.; Perkins, W.; Miller, P.S. Surgery Versus 5% Imiquimod for Nodular and Superficial Basal Cell Carcinoma: 5-Year Results of the SINS Randomized Controlled Trial. J. Investig. Dermatol. 2016, 137, 614–619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Migden, M.; Farberg, A.; Dummer, R.; Squittieri, N.; Hanke, C.W. A Review of Hedgehog Inhibitors Sonidegib and Vismodegib for Treatment of Advanced Basal Cell Carcinoma. J. Drugs Dermatol. 2021, 20, 156–165. [Google Scholar] [CrossRef]
  61. Skvara, H.; Kalthoff, F.; Meingassner, J.G.; Wolff-Winiski, B.; Aschauer, H.; Kelleher, J.F.; Wu, X.; Pan, S.; Mickel, L.; Schuster, C.; et al. Topical Treatment of Basal Cell Carcinomas in Nevoid Basal Cell Carcinoma Syndrome with a Smoothened Inhibitor. J. Investig. Dermatol. 2011, 131, 1735–1744. [Google Scholar] [CrossRef] [Green Version]
  62. Novartis Pharmaceuticals. To Evaluate the Safety, Local Tolerability, PK and PD of LDE225 on Sporadic Superficial and Nodular Skin Basal Cell Carcinomas (sBCC). ClinicalTrials.gov, 30 October 2015. Available online: https://clinicaltrials.gov/ct2/show/record/NCT01033019?term=LDE225&rank=24 (accessed on 30 May 2022).
  63. Tang, T.; Tang, J.Y.; Li, D.; Reich, M.; Callahan, C.A.; Fu, L.; Yauch, R.L.; Wang, F.; Kotkow, K.; Chang, K.S.; et al. Targeting Superficial or Nodular Basal Cell Carcinoma with Topically Formulated Small Molecule Inhibitor of Smoothened. Clin. Cancer Res. 2011, 17, 3378–3387. [Google Scholar] [CrossRef] [Green Version]
  64. Calienni, M.N.; Febres-Molina, C.; Llovera, R.E.; Zevallos-Delgado, C.; Tuttolomondo, M.E.; Paolino, D.; Fresta, M.; Barazorda-Ccahuana, H.L.; Gómez, B.; Alonso, S.D.V.; et al. Nanoformulation for potential topical delivery of Vismodegib in skin cancer treatment. Int. J. Pharm. 2019, 565, 108–122. [Google Scholar] [CrossRef] [PubMed]
  65. ClinicalTrials.gov. Clinical Trial of Patidegib Gel 2%, 4%, and Vehicle Applied Once or Twice Daily to Decrease the GLI1 Bi-omarker in Sporadic Nodular Basal Cell Carcinomas—Full Text View. Available online: https://clinicaltrials.gov/ct2/show/NCT02828111 (accessed on 11 July 2021).
  66. ClinicalTrials.gov. Trial of Patidegib Gel 2%, 4%, and Vehicle to Decrease the Number of Surgically Eligible Basal Cell Carcinomas in Gorlin Syn-drome Patients—Full Text View. Available online: https://clinicaltrials.gov/ct2/show/NCT02762084 (accessed on 11 July 2021).
  67. ClinicalTrials.gov. Study of Patidegib Topical Gel, 2%, for the Reduction of Disease Burden of Persistently Developing Basal Cell Carcinomas (BCCs) in Subjects with Basal Cell Nevus Syndrome (Gorlin Syndrome)—Full Text View. Available online: https://clinicaltrials.gov/ct2/show/NCT03703310 (accessed on 11 July 2021).
  68. Bakshi, A.; Chaudhary, S.C.; Rana, M.; Elmets, C.A.; Athar, M. Basal cell carcinoma pathogenesis and therapy involving hedgehog signaling and beyond. Mol. Carcinog. 2017, 56, 2543–2557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Kilgour, J.M.; Shah, A.; Urman, N.M.; Eichstadt, S.; Do, H.N.; Bailey, I.; Mirza, A.; Li, S.; Oro, A.E.; Aasi, S.Z.; et al. Phase II Open-Label, Single-Arm Trial to Investigate the Efficacy and Safety of Topical Remetinostat Gel in Patients with Basal Cell Carcinoma. Clin. Cancer Res. 2021, 27, 4717–4725. [Google Scholar] [CrossRef] [PubMed]
  70. Mann, B.S.; Johnson, J.R.; Cohen, M.H.; Justice, R.; Pazdur, R. FDA Approval Summary: Vorinostat for Treatment of Advanced Primary Cutaneous T-Cell Lymphoma. Oncologist 2007, 12, 1247–1252. [Google Scholar] [CrossRef] [PubMed]
  71. Zhao, J.; Quan, H.; Xie, C.; Lou, L. NL-103, a novel dual-targeted inhibitor of histone deacetylases and hedgehog pathway, effectively overcomes vismodegib resistance conferred by Smo mutations. Pharmacol. Res. Perspect. 2014, 2, e00043. [Google Scholar] [CrossRef]
  72. Kim, Y.H.; Bagot, M.; Pinter-Brown, L.; Rook, A.H.; Porcu, P.; Horwitz, S.M.; Whittaker, S.; Tokura, Y.; Vermeer, M.; Zinzani, P.L.; et al. Mogamulizumab versus vorinostat in previously treated cutaneous T-cell lymphoma (MAVORIC): An international, open-label, randomised, controlled phase 3 trial. Lancet Oncol. 2018, 19, 1192–1204. [Google Scholar] [CrossRef]
  73. Zagrodnik, B.; Kempf, W.; Seifert, B.; Müller, B.; Burg, G.; Urosevic, M.; Dummer, R. Superficial radiotherapy for patients with basal cell carcinoma. Cancer 2003, 98, 2708–2714. [Google Scholar] [CrossRef]
  74. 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] [PubMed]
  75. Avril, M.F.; Auperin, A.; Margulis, A.; Gerbaulet, A.; Duvillard, P.; Benhamou, E.; Guillaume, J.C.; Chalon, R.; Petit, J.Y.; Sancho-Garnier, H.; et al. Basal cell carcinoma of the face: Surgery or radiotherapy? Results of a randomized study. Br. J. Cancer 1997, 76, 100–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Locke, J.; Karimpour, S.; Young, G.; Lockett, M.A.; Perez, C.A. Radiotherapy for epithelial skin cancer. Int. J. Radiat. Oncol. 2001, 51, 748–755. [Google Scholar] [CrossRef]
  77. Drucker, A.M.; Adam, G.P.; Rofeberg, V.; Gazula, A.; Smith, B.; Moustafa, F.; Weinstock, M.A.; Trikalinos, T.A. Treatments of Primary Basal Cell Carcinoma of the Skin. Ann. Intern. Med. 2018, 169, 456–466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Cognetta, A.B.; Howard, B.M.; Heaton, H.P.; Stoddard, E.R.; Hong, H.G.; Green, W.H. Superficial x-ray in the treatment of basal and squamous cell carcinomas: A viable option in select patients. J. Am. Acad. Dermatol. 2012, 67, 1235–1241. [Google Scholar] [CrossRef] [PubMed]
  79. 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] [PubMed] [Green Version]
  80. Lin, C.; Tripcony, L.; Keller, J.; Poulsen, M.; Martin, J.; Jackson, J.; Dickie, G. Perineural Infiltration of Cutaneous Squamous Cell Carcinoma and Basal Cell Carcinoma Without Clinical Features. Int. J. Radiat. Oncol. 2012, 82, 334–340. [Google Scholar] [CrossRef] [PubMed]
  81. Neville, J.A.; Welch, E.; Leffell, D.J. Management of nonmelanoma skin cancer in 2007. Nat. Clin. Pract. Oncol. 2007, 4, 462–469. [Google Scholar] [CrossRef] [PubMed]
  82. Lee, C.T.; Lehrer, E.J.; Aphale, A.; Lango, M.; Galloway, T.J.; Zaorsky, N.G. Surgical excision, Mohs micrographic surgery, external-beam radiotherapy, or brachytherapy for indolent skin cancer: An international meta-analysis of 58 studies with 21,000 patients. Cancer 2019, 125, 3582–3594. [Google Scholar] [CrossRef] [PubMed]
  83. Petit, J.Y.; Avril, M.F.; Margulis, A.; Chassagne, D.; Gerbaulet, A.; Duvillard, P.; Auperin, A.; Rietjens, M. Evaluation of Cosmetic Results of a Randomized Trial Comparing Surgery and Radiotherapy in the Treatment of Basal Cell Carcinoma of the Face. Plast. Reconstr. Surg. 2000, 105, 2544–2551. [Google Scholar] [CrossRef]
  84. Peris, K.; Fargnoli, M.C.; Garbe, C.; Kaufmann, R.; Bastholt, L.; Seguin, N.B.; Bataille, V.; Marmol, V.D.; Dummer, R.; Harwood, C.A.; et al. Diagnosis and treatment of basal cell carcinoma: European consensus–based interdisciplinary guidelines. Eur. J. Cancer 2019, 118, 10–34. [Google Scholar] [CrossRef] [Green Version]
  85. Morton, C.; Dominicus, R.; Radny, P.; Dirschka, T.; Hauschild, A.; Reinhold, U.; Aschoff, R.; Ulrich, M.; Keohane, S.; Ekanayake-Bohlig, S.; et al. A randomized, multinational, noninferiority, phase III trial to evaluate the safety and efficacy of BF-200 aminolaevulinic acid gel vs. methyl aminolaevulinate cream in the treatment of nonaggressive basal cell carcinoma with photodynamic therapy. Br. J. Dermatol. 2018, 179, 309–319. [Google Scholar] [CrossRef]
  86. Fantini, F.; Greco, A.; Del Giovane, C.; Cesinaro, A.M.; Venturini, M.; Zane, C.; Surrenti, T.; Peris, K.; Calzavara-Pinton, P. Photodynamic therapy for basal cell carcinoma: Clinical and pathological determinants of response. J. Eur. Acad. Dermatol. Venereol. 2010, 25, 896–901. [Google Scholar] [CrossRef]
  87. Wang, H.; Xu, Y.; Shi, J.; Gao, X.; Geng, L. Photodynamic therapy in the treatment of basal cell carcinoma: A systematic review and meta-analysis. Photodermatol. Photoimmunol. Photomed. 2014, 31, 44–53. [Google Scholar] [CrossRef] [PubMed]
  88. Roozeboom, M.H.; Arits, A.H.H.M.; Nelemans, P.J.; Kelleners-Smeets, N.W.J. Overall treatment success after treatment of primary superficial basal cell carcinoma: A systematic review and meta-analysis of randomized and nonrandomized trials. Br. J. Dermatol. 2012, 167, 733–756. [Google Scholar] [CrossRef]
  89. Basset-Séguin, N.; Bissonnette, R.; Girard, C.; Haedersdal, M.; Lear, J.; 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. 2013, 28, 626–632. [Google Scholar] [CrossRef] [PubMed]
  90. Valladares, M.S.; Vega, J.; Prieto, M.R. Comparison of treatment of basal cell carcinoma between surgery and intralesional photodynamic therapy: A cross-sectional study. Photodiagnosis Photodyn. Ther. 2018, 21, 312–315. [Google Scholar] [CrossRef] [PubMed]
  91. Sekulic, A.; Migden, M.R.; Oro, A.E.; Dirix, L.; Lewis, K.D.; Hainsworth, J.D.; Solomon, J.A.; Yoo, S.; Arron, S.T.; Friedlander, P.A.; et al. Efficacy and Safety of Vismodegib in Advanced Basal-Cell Carcinoma. N. Engl. J. Med. 2012, 366, 2171–2179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Genentech Inc. Erivedge (vismodegib) [package insert]. U.S. Food and Drug Administration, 2012. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/203388lbl.pdf (accessed on 30 May 2022).
  93. Von Hoff, D.D.; LoRusso, P.M.; Rudin, C.M.; Reddy, J.C.; Yauch, R.L.; Tibes, R.; Weiss, G.J.; Borad, M.J.; Hann, C.L.; Brahmer, J.R.; et al. Inhibition of the Hedgehog Pathway in Advanced Basal-Cell Carcinoma. N. Engl. J. Med. 2009, 361, 1164–1172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. LoRusso, P.M.; Jimeno, A.; Dy, G.; Adjei, A.; Berlin, J.; Leichman, L.; Low, J.A.; Colburn, D.; Chang, I.; Cheeti, S.; et al. Pharmacokinetic Dose-Scheduling Study of Hedgehog Pathway Inhibitor Vismodegib (GDC-0449) in Patients with Locally Advanced or Metastatic Solid Tumors. Clin. Cancer Res. 2011, 17, 5774–5782. [Google Scholar] [CrossRef] [Green Version]
  95. Sekulic, A.; Migden, M.R.; Lewis, K.; Hainsworth, J.D.; Solomon, J.A.; Yoo, S.; Arron, S.T.; Friedlander, P.A.; Marmur, E.; Rudin, C.M.; et al. Pivotal ERIVANCE basal cell carcinoma (BCC) study: 12-month update of efficacy and safety of vismodegib in advanced BCC. J. Am. Acad. Dermatol. 2015, 72, 1021–1026.e8. [Google Scholar] [CrossRef]
  96. Dreno, B.; Basset-Seguin, N.; Caro, I.; Yue, H.; Schadendorf, D. Clinical Benefit Assessment of Vismodegib Therapy in Patients With Advanced Basal Cell Carcinoma. Oncologist 2014, 19, 790–796. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Tang, J.Y.; Mackay-Wiggan, J.M.; Aszterbaum, M.; Yauch, R.L.; Lindgren, J.; Chang, K.; Coppola, C.; Chanana, A.M.; Marji, J.; Bickers, D.R.; et al. Inhibiting the Hedgehog Pathway in Patients with the Basal-Cell Nevus Syndrome. N. Engl. J. Med. 2012, 366, 2180–2188. [Google Scholar] [CrossRef] [Green Version]
  98. Ally, M.S.; Aasi, S.; Wysong, A.; Teng, C.; Anderson, E.; Bailey-Healy, I.; Oro, A.; Kim, J.; Chang, A.L.; Tang, J.Y. An investigator-initiated open-label clinical trial of vismodegib as a neoadjuvant to surgery for high-risk basal cell carcinoma. J. Am. Acad. Dermatol. 2014, 71, 904–911.e1. [Google Scholar] [CrossRef]
  99. Novartis Pharmaceuticals. Odomzo (Sonidegib) [Package Insert]. U.S. Food and Drug Administration, 24 July 2015. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2015/205266Orig1s000lbl.pdf (accessed on 30 May 2022).
  100. Pan, S.; Wu, X.; Jiang, J.; Gao, W.; Wan, Y.; Cheng, D.; Han, D.; Liu, J.; Englund, N.P.; Wang, Y.; et al. Discovery of NVP-LDE225, a Potent and Selective Smoothened Antagonist. ACS Med. Chem. Lett. 2010, 1, 130–134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Doan, H.Q.; Silapunt, S.; Migden, M.R. Sonidegib, a novel smoothened inhibitor for the treatment of advanced basal cell carcinoma. OncoTargets Ther. 2016, ume 9, 5671–5678. [Google Scholar] [CrossRef] [Green Version]
  102. Rodon, J.; Tawbi, H.A.; Thomas, A.L.; Stoller, R.G.; Turtschi, C.P.; Baselga, J.; Sarantopoulos, J.; Mahalingam, D.; Shou, Y.; Moles, M.A.; et al. A Phase I, Multicenter, Open-Label, First-in-Human, Dose-Escalation Study of the Oral Smoothened Inhibitor Sonidegib (LDE225) in Patients with Advanced Solid Tumors. Clin. Cancer Res. 2014, 20, 1900–1909. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Migden, M.R.; Guminski, A.; Gutzmer, R.; Dirix, L.; Lewis, K.D.; Combemale, P.; Herd, R.M.; Kudchadkar, R.; Trefzer, U.; Gogov, S.; et al. Treatment with two different doses of sonidegib in patients with locally advanced or metastatic basal cell carcinoma (BOLT): A multicentre, randomised, double-blind phase 2 trial. Lancet Oncol. 2015, 16, 716–728. [Google Scholar] [CrossRef]
  104. Dummer, R.; Guminksi, A.; Gutzmer, R.; Lear, J.; Lewis, K.; Chang, A.; Combemale, P.; Dirix, L.; Kaatz, M.; Kudchadkar, R.; et al. Long-term efficacy and safety of sonidegib in patients with advanced basal cell carcinoma: 42-month analysis of the phase II randomized, double-blind BOLT study. Br. J. Dermatol. 2019, 182, 1369–1378. [Google Scholar] [CrossRef] [PubMed]
  105. 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] [PubMed] [Green Version]
  106. Stratigos, A.J.; Sekulic, A.; Peris, K.; Bechter, O.; Prey, S.; Kaatz, M.; Lewis, K.D.; Basset-Seguin, N.; Chang, A.L.S.; Dalle, S.; et al. Cemiplimab in locally advanced basal cell carcinoma after hedgehog inhibitor therapy: An open-label, multi-centre, single-arm, phase 2 trial. Lancet Oncol. 2021, 22, 848–857. [Google Scholar] [CrossRef]
  107. Damsin, T.; Lebas, E.; Marchal, N.; Rorive, A.; Nikkels, A.F. Cemiplimab for locally advanced and metastatic basal cell carcinoma. Expert Rev. Anticancer Ther. 2022, 22, 243–248. [Google Scholar] [CrossRef] [PubMed]
  108. Davis, C.M.; Lewis, K.D. Brief overview: Cemiplimab for the treatment of advanced basal cell carcinoma: PD-1 strikes again. Ther. Adv. Med Oncol. 2022, 14, 17588359211066147. [Google Scholar] [CrossRef]
  109. Jin, G.; Sivaraman, A.; Lee, K. Development of taladegib as a sonic hedgehog signaling pathway inhibitor. Arch. Pharmacal Res. 2017, 40, 1390–1393. [Google Scholar] [CrossRef] [PubMed]
  110. Bendell, J.; Andre, V.; Ho, A.; Kudchadkar, R.; Migden, M.; Infante, J.; Tiu, R.V.; Pitou, C.; Tucker, T.; Brail, L.; et al. Phase I Study of LY2940680, a Smo Antagonist, in Patients with Advanced Cancer Including Treatment-Naïve and Previously Treated Basal Cell Carcinoma. Clin. Cancer Res. 2018, 24, 2082–2091. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Jimeno, A.; Weiss, G.J.; Miller, W.H.; Gettinger, S.; Eigl, B.J.; Chang, A.L.S.; Dunbar, J.; Devens, S.; Faia, K.; Skliris, G.; et al. Phase I Study of the Hedgehog Pathway Inhibitor IPI-926 in Adult Patients with Solid Tumors. Clin. Cancer Res. 2013, 19, 2766–2774. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. ClinicalTrials.gov. Extension Study of Patidegib Topical Gel, 2% in Subjects with Gorlin Syndrome (Basal Cell Nevus Syn-drome)—Full Text View. Available online: https://clinicaltrials.gov/ct2/show/NCT04308395 (accessed on 11 July 2021).
  113. Novartis Pharmaceuticals. A Phase I, Multi-Center, Open Label, Dose Escalation Study of LEQ506, an Oral Smoothened Inhibitor, in Patients with Advanced Solid Tumors. clinicaltrials.gov; Clinical Trial Registration NCT01106508; December 2020. Available online: https://clinicaltrials.gov/ct2/show/NCT01106508 (accessed on 26 May 2022).
  114. Peukert, S.; He, F.; Dai, M.; Zhang, R.; Sun, Y.; Miller-Moslin, K.; McEwan, M.; Lagu, B.; Wang, K.; Yusuff, N.; et al. Discovery of NVP-LEQ506, a Second-Generation Inhibitor of Smoothened. ChemMedChem 2013, 8, 1261–1265. [Google Scholar] [CrossRef] [PubMed]
  115. Lauressergues, E.; Heusler, P.; Lestienne, F.; Troulier, D.; Rauly-Lestienne, I.; Tourette, A.; Ailhaud, M.; Cathala, C.; Tardif, S.; Denais-Laliève, D.; et al. Pharmacological evaluation of a series of smoothened antagonists in signaling pathways and after topical application in a depilated mouse model. Pharmacol. Res. Perspect. 2016, 4, e00214. [Google Scholar] [CrossRef]
  116. Guangdong Zhongsheng Pharmaceutical Co., Ltd. A Phase 1, Open-Label, Dose-Escalation and Expansion, Safety and Tolerability Study of ZSP1602 in Participants with Advanced Solid Tumors. clinicaltrials.gov; Clinical Trial Registration NCT03734913; July 2020. Available online: https://clinicaltrials.gov/ct2/show/NCT03734913 (accessed on 26 May 2022).
  117. Jia, J. Decoding the Hedgehog signal in animal development. Experientia 2006, 63, 1249–1265. [Google Scholar] [CrossRef]
  118. Jia, H.; Liu, Y.; Xia, R.; Tong, C.; Yue, T.; Jiang, J.; Jia, J. Casein Kinase 2 Promotes Hedgehog Signaling by Regulating both Smoothened and Cubitus Interruptus. J. Biol. Chem. 2010, 285, 37218–37226. [Google Scholar] [CrossRef] [Green Version]
  119. Giroux-Leprieur, E.; Costantini, A.; Ding, V.W.; He, B. Hedgehog Signaling in Lung Cancer: From Oncogenesis to Cancer Treatment Resistance. Int. J. Mol. Sci. 2018, 19, 2835. [Google Scholar] [CrossRef] [Green Version]
  120. Zhang, S.; Wang, Y.; Mao, J.-H.; Hsieh, D.; Kim, I.-J.; Hu, L.-M.; Xu, Z.; Long, H.; Jablons, D.M.; You, L. Inhibition of CK2α Down-Regulates Hedgehog/Gli Signaling Leading to a Reduction of a Stem-Like Side Population in Human Lung Cancer Cells. PLoS ONE 2012, 7, e38996. [Google Scholar] [CrossRef] [Green Version]
  121. Purzner, T.; Purzner, J.; Buckstaff, T.; Cozza, G.; Gholamin, S.; Rusert, J.M.; Hartl, T.A.; Sanders, J.; Conley, N.; Ge, X.; et al. Developmental phosphoproteomics identifies the kinase CK2 as a driver of Hedgehog signaling and a therapeutic target in medulloblastoma. Sci. Signal. 2018, 11, eaau5147. [Google Scholar] [CrossRef] [Green Version]
  122. Senhwa Biosciences, Inc. A Phase I, Multi-Center, Open-Label, Treatment Duration Increment, Expansion, Safety, and Pharmacodynamic Study of CX-4945 Administered Orally Twice Daily to Patients with Advanced Basal Cell Carcinoma. clinicaltrials.gov; Clinical Trial Registration NCT03897036; March 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT03897036 (accessed on 26 May 2022).
  123. Kim, J.; Tang, J.Y.; Gong, R.; Kim, J.; Lee, J.J.; Clemons, K.V.; Chong, C.R.; Chang, K.S.; Fereshteh, M.; Gardner, D.; et al. Itraconazole, a Commonly Used Antifungal that Inhibits Hedgehog Pathway Activity and Cancer Growth. Cancer Cell 2010, 17, 388–399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Fry, M.; Teng, C.; Lee, A.; Sun, W.; Parekh, M.; Rajadas, J.; Beachy, P.; Epstein, E.; Tang, J. LB783 Topical Itraconazole inhibits the Hedgehog signaling pathway and reduces tumor development and size in murine models. J. Investig. Dermatol. 2016, 136, B6. [Google Scholar] [CrossRef]
  125. Kim, D.J.; Kim, J.; Spaunhurst, K.; Montoya, J.; Khodosh, R.; Chandra, K.; Fu, T.; Gilliam, A.; Molgo, M.; Beachy, P.A.; et al. Open-Label, Exploratory Phase II Trial of Oral Itraconazole for the Treatment of Basal Cell Carcinoma. J. Clin. Oncol. 2014, 32, 745–751. [Google Scholar] [CrossRef]
  126. Sohn, G.K.; Kwon, G.P.; Bailey-Healy, I.; Mirza, A.; Sarin, K.; Oro, A.; Tang, J.Y. Topical Itraconazole for the Treatment of Basal Cell Carcinoma in Patients With Basal Cell Nevus Syndrome or High-Frequency Basal Cell Carcinomas. JAMA Dermatol. 2019, 155, 1078–1080. [Google Scholar] [CrossRef]
  127. Ally, M.S.; Ransohoff, K.J.; Sarin, K.; Atwood, S.; Rezaee, M.; Bailey-Healy, I.; Kim, J.; Beachy, P.A.; Chang, A.L.S.; Oro, A.E.; et al. Effects of Combined Treatment With Arsenic Trioxide and Itraconazole in Patients With Refractory Metastatic Basal Cell Carcinoma. JAMA Dermatol. 2016, 152, 452–456. [Google Scholar] [CrossRef] [PubMed]
  128. Kasper, M.; Regl, G.; Frischauf, A.-M.; Aberger, F. GLI transcription factors: Mediators of oncogenic Hedgehog signalling. Eur. J. Cancer 2006, 42, 437–445. [Google Scholar] [CrossRef] [PubMed]
  129. Ingham, P.W.; Nakano, Y.; Seger, C. Mechanisms and functions of Hedgehog signalling across the metazoa. Nat. Rev. Genet. 2011, 12, 393–406. [Google Scholar] [CrossRef] [PubMed]
  130. Hui, C.-C.; Angers, S. Gli Proteins in Development and Disease. Annu. Rev. Cell Dev. Biol. 2011, 27, 513–537. [Google Scholar] [CrossRef] [Green Version]
  131. Lauth, M.; Bergström, Å.; Shimokawa, T.; Toftgård, 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] [Green Version]
  132. Wickström, M.; Dyberg, C.; Shimokawa, T.; Milosevic, J.; Baryawno, N.; Fuskevåg, O.M.; Larsson, R.; Kogner, P.; Zaphiropoulos, P.G.; Johnsen, J.I. Targeting the hedgehog signal transduction pathway at the level of GLI inhibits neuroblastoma cell growth in vitro and in vivo. Int. J. Cancer 2012, 132, 1516–1524. [Google Scholar] [CrossRef]
  133. Chen, Q.; Xu, R.; Zeng, C.; Lu, Q.; Huang, D.; Shi, C.; Zhang, W.; Deng, L.; Yan, R.; Rao, H.; et al. 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] [PubMed] [Green Version]
  134. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Faniku, C.; Kong, W.; He, L.; Zhang, M.; Lilly, G.; Pepper, J. Hedgehog signaling promotes endoneurial fibroblast migration and Vegf-A expression following facial nerve injury. Brain Res. 2020, 1751, 147204. [Google Scholar] [CrossRef] [PubMed]
  136. Johnson, K.E.; Wilgus, T.A. Multiple Roles for VEGF in Non-Melanoma Skin Cancer: Angiogenesis and Beyond. J. Ski. Cancer 2012, 2012, 1–6. [Google Scholar] [CrossRef] [PubMed]
  137. AiViva BioPharma, Inc. An Exploratory Study to Evaluate the Safety and Efficacy of AIV001 Intradermally Administered in Subjects with Biopsy-Confirmed Basal Cell Carcinoma. clinicaltrials.gov; Clinical Trial Registration NCT04470726; May 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT04470726 (accessed on 26 May 2022).
  138. Massagué, J. TGFβ signalling in context. Nat. Rev. Mol. Cell Biol. 2012, 13, 616–630. [Google Scholar] [CrossRef]
  139. Kuonen, F.; Surbeck, I.; Sarin, K.Y.; Dontenwill, M.; Rüegg, C.; Gilliet, M.; Oro, A.E.; Gaide, O. TGFβ, Fibronectin and Integrin α5β1 Promote Invasion in Basal Cell Carcinoma. J. Investig. Dermatol. 2018, 138, 2432–2442. [Google Scholar] [CrossRef] [Green Version]
  140. Chen, Y.; Liu, J. The prognostic roles of cyclooxygenase-2 for patients with basal cell carcinoma. Artif. Cells Nanomed. Biotechnol. 2019, 47, 3053–3057. [Google Scholar] [CrossRef] [Green Version]
  141. El-Khalawany, M.A.; Abou-Bakr, A.A. Role of Cyclooxygenase-2, Ezrin and Matrix metalloproteinase-9 as predictive markers for recurrence of basal cell carcinoma. J. Cancer Res. Ther. 2013, 9, 613–617. [Google Scholar] [CrossRef]
  142. Sirnaomics. An Open Label, Dose Escalation Study to Evaluate the Safety and Efficacy of Localized Injection of STP705 in Adult Patients with Basal Cell Carcinoma. clinicaltrials.gov; Clinical Trial Registration NCT04669808; January 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT04669808 (accessed on 30 May 2022).
  143. Grosso, J.F.; Goldberg, M.V.; Getnet, D.; Bruno, T.C.; Yen, H.-R.; Pyle, K.J.; Hipkiss, E.; Vignali, D.A.A.; Pardoll, D.M.; Drake, C.G. Functionally Distinct LAG-3 and PD-1 Subsets on Activated and Chronically Stimulated CD8 T Cells. J. Immunol. 2009, 182, 6659–6669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Yang, Z.-Z.; Kim, H.J.; Villasboas, J.C.; Chen, Y.-P.; Price-Troska, T.; Jalali, S.; Wilson, M.; Novak, A.J.; Ansell, S.M. Expression of LAG-3 defines exhaustion of intratumoral PD-1+ T cells and correlates with poor outcome in follicular lymphoma. Oncotarget 2017, 8, 61425–61439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Tawbi, H.A.; Schadendorf, D.; Lipson, E.J.; Ascierto, P.A.; Matamala, L.; Gutiérrez, E.C.; Rutkowski, P.; Gogas, H.J.; Lao, C.D.; De Menezes, J.J.; et al. Relatlimab and Nivolumab versus Nivolumab in Untreated Advanced Melanoma. N. Engl. J. Med. 2022, 386, 24–34. [Google Scholar] [CrossRef] [PubMed]
  147. Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins. Nivolumab Alone or Plus Relatlimab or Ipilimumab for Patients with Locally-Advanced Unresectable or Metastatic Basal Cell Carcinoma. clinicaltrials.gov; Clinical Trial Registration NCT03521830; March 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT03521830 (accessed on 26 May 2022).
  148. Ferrucci, P.; Pala, L.; Conforti, F.; Cocorocchio, E. Talimogene Laherparepvec (T-VEC): An Intralesional Cancer Immunotherapy for Advanced Melanoma. Cancers 2021, 13, 1383. [Google Scholar] [CrossRef] [PubMed]
  149. Blackmon, J.T.; Dhawan, R.; Viator, T.M.; Terry, N.L.; Conry, R.M. Talimogene laherparepvec for regionally advanced Merkel cell carcinoma: A report of 2 cases. JAAD Case Rep. 2017, 3, 185–189. [Google Scholar] [CrossRef] [Green Version]
  150. Ramelyte, E.; Tastanova, A.; Balázs, Z.; Ignatova, D.; Turko, P.; Menzel, U.; Guenova, E.; Beisel, C.; Krauthammer, M.; Levesque, M.P.; et al. Oncolytic virotherapy-mediated anti-tumor response: A single-cell perspective. Cancer Cell 2021, 39, 394–406.e4. [Google Scholar] [CrossRef]
  151. University of Zurich. A Phase I, Open Label, Single Arm, Single Centre Study to Evaluate Mechanism of Action of Talimogene Laherparepvec (T-VEC) in Locally Advanced Non-Melanoma Skin Cancer. clinicaltrials.gov; Clinical Trial Registration NCT03458117; March 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT03458117 (accessed on 26 May 2022).
  152. National Cancer Institute (NCI). A Phase II Study of Talimogene Laherparepvec Followed by Talimogene Laherparepvec + Nivolumab in Refractory T Cell and NK Cell Lymphomas, Cutaneous Squamous Cell Carcinoma, Merkel Cell Carcinoma, and Other Rare Skin Tumors. clinicaltrials.gov; Clinical Trial Registration NCT02978625; April 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT02978625 (accessed on 26 May 2022).
  153. Miura, J.T.; Zager, J.S. Intralesional therapy as a treatment for locoregionally metastatic melanoma. Expert Rev. Anticancer Ther. 2018, 18, 399–408. [Google Scholar] [CrossRef]
  154. Wilmas, K.M.; Nguyen, Q.-B.; Patel, J.; Silapunt, S.; Migden, M.R. Treatment of advanced cutaneous squamous cell carcinoma: A Mohs surgery and dermatologic oncology perspective. Futur. Oncol. 2021, 17, 4971–4982. [Google Scholar] [CrossRef]
  155. Zawit, M.; Swami, U.; Awada, H.; Arnouk, J.; Milhem, M.; Zakharia, Y. Current status of intralesional agents in treatment of malignant melanoma. Ann. Transl. Med. 2021, 9, 1038. [Google Scholar] [CrossRef]
  156. Van Horssen, R.; Hagen, T.L.M.T.; Eggermont, A.M.M. TNF-α in Cancer Treatment: Molecular Insights, Antitumor Effects, and Clinical Utility. Oncologist 2006, 11, 397–408. [Google Scholar] [CrossRef]
  157. Malek, T.R. The main function of IL-2 is to promote the development of T regulatory cells. J. Leukoc. Biol. 2003, 74, 961–965. [Google Scholar] [CrossRef]
  158. Philogen, S.p.A. A Phase II Study of Intratumoral Administration of L19IL2/L19TNF in Non-melanoma Skin Cancer Patients with Presence of Injectable Lesions. clinicaltrials.gov; Clinical Trial Registration NCT04362722; April 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT04362722 (accessed on 26 May 2022).
  159. Danielli, R.; Patuzzo, R.I.; Di Giacomo, A.M.; Gallino, G.; Maurichi, A.; Di Florio, A.; Cutaia, O.; Lazzeri, A.; Fazio, C.; Miracco, C.; et al. Intralesional administration of L19-IL2/L19-TNF in stage III or stage IVM1a melanoma patients: Results of a phase II study. Cancer Immunol. Immunother. 2015, 64, 999–1009. [Google Scholar] [CrossRef] [PubMed]
  160. Ni, L.; Lu, J. Interferon gamma in cancer immunotherapy. Cancer Med. 2018, 7, 4509–4516. [Google Scholar] [CrossRef] [PubMed]
  161. Wang, Q.-S.; Shen, S.-Q.; Sun, H.-W.; Xing, Z.-X.; Yang, H.-L. Interferon-gamma induces autophagy-associated apoptosis through induction of cPLA2-dependent mitochondrial ROS generation in colorectal cancer cells. Biochem. Biophys. Res. Commun. 2018, 498, 1058–1065. [Google Scholar] [CrossRef]
  162. Kammertoens, T.; Friese, C.; Arina, A.; Idel, C.; Briesemeister, D.; Rothe, M.; Ivanov, A.; Szymborska-Mell, A.; Patone, G.; Kunz, S.; et al. Tumour ischaemia by interferon-γ resembles physiological blood vessel regression. Nature 2017, 545, 98–102. [Google Scholar] [CrossRef] [PubMed]
  163. Aqbi, H.F.; Wallace, M.; Sappal, S.; Payne, K.K.; Manjili, M.H. IFN-γ orchestrates tumor elimination, tumor dormancy, tumor escape, and progression. J. Leukoc. Biol. 2018, 103, 1219–1223. [Google Scholar] [CrossRef] [PubMed]
  164. Ascend Biopharmaceuticals Ltd. A Phase 2A Study to Assess the Safety and Efficacy of ASN-002 Combined With a Hedgehog Pathway Inhibitor in the Treatment of Multiple Low Risk Basal Cell Carcinomas in Sporadic or Basal Cell Nevus Syndrome Patients. clinicaltrials.gov; Clinical Trial Registration NCT04416516; February 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT04416516 (accessed on 26 May 2022).
Table 1. Treatments for consideration by category for BCCs with lower or higher-risk features.
Table 1. Treatments for consideration by category for BCCs with lower or higher-risk features.
Treatments for
Consideration by Category 1
BCCs with Lower-Risk FeaturesBCCs with Higher-Risk Features
Procedural
treatments
Standard Excision with postoperative margin assessment
Electrodesiccation and curettage
Cryotherapy
Neodymium-doped yttrium aluminum garnet (Nd:YAG) laser ablation.
Carbon-dioxide laser ablation
Mohs micrographic surgery
Standard Excision with postoperative margin assessment
Topical,
Intralesional, and Field Treatments
Topical imiquimod
Topical 5-fluorouracil
Topical remetinostat (HDAC inhibitor)
Photodynamic therapy
Radiation Therapy
Radiation therapy
Systemic
Treatments
Vismodegib
Sonidegib
Cemiplimab
Investigational Treatments 2
Patidegib/TAK-441 (SMO inhibitor, topical)
AIV001/Anti-VEGFR
IL-2/TNF-α (intralesional)
IFN gamma adenovirus/ASN-002 (intralesional)
Taladegib/LY2940680 (SMO inhibitor)
LEQ506 (SMO inhibitor)
ZSP 1602 (SMO inhibitor)
CX-4945 (CK2 Inhibitor)
Itraconazole (SMO inhibitor- combined with vismodegib or sonidegib or arsenic trioxide)
Anti LAG3 Ab
Intralesional talimogene laherparepvec (T-VEC)
Anti COX-2 TGF-β siRNA
IL-2/TNF-α (intralesional)
IFN gamma adenovirus/ASN-002 (intralesional)
Vorinostat (HDAC inhibitor)
1 Treatments are listed for consideration within the comprehensive clinicopathologic context of the patient under evaluation for treatment. 2 Some treatments discussed in this review are excluded from the table because there is no clinically available medication for 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

Chen, O.M.; Kim, K.; Steele, C.; Wilmas, K.M.; Aboul-Fettouh, N.; Burns, C.; Doan, H.Q.; Silapunt, S.; Migden, M.R. Advances in Management and Therapeutics of Cutaneous Basal Cell Carcinoma. Cancers 2022, 14, 3720. https://doi.org/10.3390/cancers14153720

AMA Style

Chen OM, Kim K, Steele C, Wilmas KM, Aboul-Fettouh N, Burns C, Doan HQ, Silapunt S, Migden MR. Advances in Management and Therapeutics of Cutaneous Basal Cell Carcinoma. Cancers. 2022; 14(15):3720. https://doi.org/10.3390/cancers14153720

Chicago/Turabian Style

Chen, Olivia M., Keemberly Kim, Chelsea Steele, Kelly M. Wilmas, Nader Aboul-Fettouh, Carrick Burns, Hung Quoc Doan, Sirunya Silapunt, and Michael R. Migden. 2022. "Advances in Management and Therapeutics of Cutaneous Basal Cell Carcinoma" Cancers 14, no. 15: 3720. https://doi.org/10.3390/cancers14153720

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