Next Article in Journal
The Renoprotective Mechanisms of Sodium-Glucose Cotransporter-2 Inhibitors (SGLT2i)—A Narrative Review
Previous Article in Journal
Proteomic Analysis of Human Macrophages Overexpressing Angiotensin-Converting Enzyme
Previous Article in Special Issue
The Effects of ML385 on Head and Neck Squamous Cell Carcinoma: Implications for NRF2 Inhibition as a Therapeutic Strategy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Approaches for Non-Melanoma Skin Cancer: Standard of Care and Emerging Modalities

by
Stefano Sol
1,
Fabiana Boncimino
1,
Kristina Todorova
1,
Sarah Elizabeth Waszyn
2 and
Anna Mandinova
1,3,4,*
1
Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
2
Drexel University College of Medicine, 60 N 36th Street, Philadelphia, PA 19104, USA
3
Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, MA 02142, USA
4
Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(13), 7056; https://doi.org/10.3390/ijms25137056
Submission received: 22 May 2024 / Revised: 20 June 2024 / Accepted: 22 June 2024 / Published: 27 June 2024
(This article belongs to the Special Issue Advances in Head and Neck and Skin Cancer)

Abstract

:
Skin cancer encompasses a range of cutaneous malignancies, with non-melanoma skin cancers (NMSCs) being the most common neoplasm worldwide. Skin exposure is the leading risk factor for initiating NMSC. Ultraviolet (UV) light induces various genomic aberrations in both tumor-promoting and tumor-suppressing genes in epidermal cells. In conjunction with interactions with a changed stromal microenvironment and local immune suppression, these aberrations contribute to the occurrence and expansion of cancerous lesions. Surgical excision is still the most common treatment for these lesions; however, locally advanced or metastatic disease significantly increases the chances of morbidity or death. In recent years, numerous pharmacological targets were found through extensive research on the pathogenic mechanisms of NMSCs, leading to the development of novel treatments including Hedgehog pathway inhibitors for advanced and metastatic basal cell carcinoma (BCC) and PD-1/PD-L1 inhibitors for locally advanced cutaneous squamous cell carcinoma (cSCC) and Merkel cell carcinoma (MCC). Despite the efficacy of these new drugs, drug resistance and tolerability issues often arise with long-term treatment. Ongoing studies aim to identify alternative strategies with reduced adverse effects and increased tolerability. This review summarizes the current and emerging therapies used to treat NMSC.

1. Introduction

Skin cancer has the highest incidence of all forms of cancer worldwide, with approximately 90% of these cases classified as non-melanoma skin cancer (NMSC) [1]. The leading cause of NMSCs is unprotected exposure to ultraviolet (UV) radiation, although many other variables also contribute to their pathogenesis [2]. Among these factors, immune suppression from various causes significantly exacerbates the risk and aggressiveness of NMSCs. In 2020, GLOBOCAN estimated that approximately 1.2 million new cases of NMSC occur annually, though this count may be underestimated due to challenges with NMSC identification and documentation [3]. Among NMSCs, the most frequent are basal cell carcinoma (BCC) and cutaneous squamous cell carcinoma (cSCC), while Merkel cell carcinoma (MCC) is less commonly encountered [4]. While the mortality rate from these cancers is relatively low, the prognosis worsens if the cancers metastasize. The incidence of metastatic BCC is estimated to be less than 0.1%, with a median overall survival (OS) of 10 months [5]. The metastatic potential of cSCC varies from 0.3% to 3.7% with a median OS of 2.19 years [6]. cSCC and BCC have better prognoses than MCC, which has a 5-year OS rate of ≤18% [7].
Surgical excision is still the most prevalent therapeutic approach in individuals who present with NMSC [8]. Late-stage NMSC patients, however, often are not candidates for surgery and have significant rates of post-surgical recurrence if they are surgical candidates. Recent studies aim to solve this problem by developing targeted therapeutics and immunotherapy [9]. Researchers have identified dysregulated intracellular signaling pathways in NMSCs for this purpose. The hedgehog protein signaling pathway has been identified as a target for BCC, and the epidermal growth factor receptor (EGFR) has emerged as a target for cSCC. Immunotherapy studies have progressed in treating systemic disease by applying immuno-checkpoint inhibitors targeting the PD-1/PD-L1 pathway and CTLA-4. The development of treatments with the highest efficacy and fewest adverse effects will necessitate continued research for years to come. This review aims to provide a comprehensive exploration of the traditional and the latest therapeutic approaches in the treatment of NMSCs.

2. Pathogenesis of NMSC

NMSCs are characterized by aberrant cell growth and a high mutational burden. Their development is primarily due to exposure to ultraviolet radiation (UV), although fair skin, immunosuppression, viruses, and certain hereditary disorders also contribute to the pathogenesis of these diseases [10,11].

2.1. UV Radiation

The most prevalent sites of NMSC are the most sun-exposed areas of the body, such as the head and neck region, followed by the upper limbs [12,13]. A correlation between the geographical incidence of NMSC and latitudinal variation in the intensities of UV radiation has been demonstrated [12,14]. Countries with the highest risk of NMSC incidence are predominantly those with a more significant proportion of people with fair skin [1]. Individuals with darker skin tones, who have higher melanin content, are generally more protected against UV-induced damage [15]. UV radiation exerts its destructive effects through DNA damage and oxidative stress, which can subsequently lead to gene mutations and inflammatory responses [16,17]. The dysregulation of diverse signaling pathways caused by UV damage has been shown to impair the ability to metabolize free radicals and to promote abnormal keratinocyte mitosis. These changes may contribute to the development of NMSC [18].

2.2. Immunosuppression

Individuals with impaired immune systems are at a relatively increased risk of developing cutaneous malignancies when compared to healthy individuals [11]. Immunosuppressed patient cohorts comprise solid organ transplant recipients, patients afflicted with hematologic neoplasms such as chronic lymphocytic leukemia, and individuals suffering from certain viral infectious diseases such as acquired immunodeficiency syndrome (AIDS) [19,20]. Patients who have undergone transplantation and thus have been treated with prolonged immunosuppressive therapy exhibit an increased risk of developing skin malignancies [21]. Among the NMSC, the incidence of cSCC in immunosuppressed patients is the most prominent [22,23]. Furthermore, tumors exhibit more aggressive behavior in transplant recipients, with a higher risk of metastasis and mortality [24].

2.3. Oncogenic and Tumor Suppressive Pathway

Compared to all other types of cancer, NMSCs are associated with a significantly elevated tumor mutational burden (TMB). The median number of mutations per megabase (Mb) is 45.2, 47.3, and 53.9 in cSCC, BCC, and MCC, respectively [25,26]. The high TMB is thought to be related to prolonged exposure to UV radiation. The mutational landscape of NMSC typically results in the impairment of signal transduction pathways involved in cell cycle control and the regulation of differentiation [27,28].
Various mutations are detected in cSCC, most commonly in the TP53, NOTCH1, NOTCH2, CDKN2A, HRAS, and EGFR genes [27,29]. Mutations in the tumor suppressor p53 can lead to alterations in the apoptosis of cells with damaged DNA and uncontrolled proliferation of keratinocytes [30,31]. Meanwhile, mutations inhibiting NOTCH signaling, often in combination with RAS and PI3K mutations, are primarily associated with the dysregulation of keratinocyte differentiation [32,33]. Targeted therapeutic strategies mainly focus on inhibiting the EGFR, which is frequently mutated or deregulated in cSCC [34]. Its dysregulation leads to downstream activation of signaling pathways, including Ras-Raf-MEK-ERK and PI3K, which affect keratinocyte proliferation and differentiation [34].
The Hedgehog (Hh) signaling pathway is central to BCC initiation and progression [35]. Dysregulated activation of the Hh signaling pathway is mainly due to Patched 1 (PTCH1) loss-of-function mutations and G-protein-coupled smoothed (SMO) transmembrane receptor activating mutations [36]. PTCH1 is a constitutive inhibitor of SMO. In the presence of the ligand Sonic Hedgehog, SMO is no longer repressed by PTCH1, activating the SMO-SUFU-GLI cascade. Glioma-associated (GLI) transcription factors are free to translocate to the nucleus, where they regulate the transcription of several genes that control cell proliferation and growth [37].
By contrast, two pathways are thought to contribute to MCC pathogenesis: UV radiation and Merkel cell polyomavirus (MPyV) [38]. The development of MCC is more often associated with MPyV [39]. Virus-positive (VP)-MCC accounts for almost 80% of the MCCs in the USA, while the incidence of virus-negative (VN)-MCC increases in areas with high UV exposure and fair-skinned populations [40,41]. It is important to note that VP- and VN-MCC show significant differences in mutational burden and genomic stability, with TP53 and RB1 being the most frequently mutated genes in VN-MCC [26,42]. Meanwhile, small T antigen (sT) and the truncated form of large T antigen (LT), which are expressed upon the integration of MPyV DNA into the host cells, are critical drivers for carcinogenesis in VP-MCC [43]. The essential function of truncated LT in VP-MCC is to inhibit Rb and p53, which, similarly to VN-MCC, leads to uncontrolled MCC cell proliferation [44,45].

3. Cutaneous Squamous Cell Carcinoma

3.1. Clinical Features and Staging of cSCC

cSCC often appears as a non-healing ulcer or an irregular growth on sun-exposed body parts, such as the head, neck, lower lip, scalp, and forehead. It typically manifests as nodules or plaques characterized by varying degrees of ulceration, crusting, and scaling [46]. The anatomical location and subtype impact the highly variable clinical characteristics of cSCC [47,48]. These subtypes differ significantly in their biological behavior, with some showing a strong predisposition for aggressive and often metastatic activity and others being relatively indolent. Several cSCC subtypes can be categorized as low-, intermediate-, or high-risk variations based on the likelihood of metastasis and tumor recurrence [49]. cSCC in situ lesions are the earliest form of squamous cell skin cancer. They usually grow slowly and present with no symptoms [46]. These kinds of lesions can vary in appearance from a tiny keratotic papule to a scaly pink area. A clearly defined, erythematous, red plaque is the hallmark of Bowen disease, a form of in situ cSCC [50]. Invasive cSCC may develop from up to 5% of in situ cSCC. Histologic perineural invasion is seen in lesions of invasive cSCC, particularly in high-risk cases [51].
cSCC presents a challenge in identifying high-risk cases, complicating treatment strategies and prognosis assessment. Other factors, such as nodal involvement and in-transit metastases, further add to the complexity of prognosis determination [52,53]. Brigham and Women’s Hospital (BWH) (Boston, MA, USA) has identified four key risk factors that improve predictive accuracy compared to traditional staging systems like the American Joint Committee on Cancer (AJCC) and Union for International Cancer Control (UICC) [54]. These include tumor diameter ≥ 2 cm (linked to local recurrence), invasion beyond subcutaneous fat (associated with nodal metastasis risk), poor differentiation status, and perineural invasion (the latter two both being related to disease-specific death risk). The National Comprehensive Cancer Network (NCCN) defines high-risk cSCC based on various clinical and histological factors, including tumor size, margin definition, recurrence, and histological characteristics like poor differentiation [55]. The NCCN mainly focuses on treatment guidance, while the BWH prioritizes prognostic assessment, and the AJCC categorizes stages from T0 to T4 based on tumor size and depth of invasion [56]. Recent studies have demonstrated that BWH predicts metastasis and mortality more effectively than AJCC [57]. This is due to its higher specificity and positive predictive value. This classification framework assists clinicians in devising treatment plans and forecasting patient outcomes [49,58].

3.2. Current Available Therapies

cSCC is predominantly managed through surgical excision and histopathologic scrutiny, achieving remarkable cure rates of up to 95% [4]. Initial management of early-stage cSCC predominantly relies on surgical resection, boasting a meager recurrence rate of approximately 4.6% and negligible metastatic potential [59,60]. High-risk cSCCs are best managed with micrographically controlled surgery (MCS), which minimizes the removal of uninvolved tissue and provides a thorough margin inspection [61].
In scenarios where surgery is not indicated, radiation therapy emerges as a viable alternative. Radiotherapy offers compelling outcomes, showcasing high cure rates particularly in early-stage cSCC [62,63]. Although these standard-of-care methods are effective, they are often invasive and may lead to considerable scarring and fibrosis. Alternatively, the management of cSCC includes various physical treatment options such as laser therapies, cryotherapy, curettage, and photodynamic therapy [64,65,66]. Among these, photodynamic therapy (PDT) utilizes a photosensitizer along with light exposure in an oxygen-rich environment to eliminate cancer cells. Over the past twenty years, PDT has developed into an effective treatment method for NMSCs including superficial and thin nodular BBC and in situ cSCC.
These modalities are highly effective for low-risk, localized cSCC, especially when conventional surgical intervention is not an option. Despite their effectiveness and favorable aesthetic outcomes, these modalities lack histological assessment capabilities. Advanced cSCC, typified by stage III or higher tumors, poses therapeutic challenges that extend beyond conventional surgical or radiotherapeutic interventions.
Figure 1 illustrates the algorithm for the treatment of cSCC.

3.2.1. Topical Treatment

For patients who are not candidates for surgery, topical treatments such as imiquimod or 5-fluorouracil (5-FU) creams offer alternative approaches to manage precancerous lesions or localized cSCC. Topical 5-FU disrupts intracellular nucleotide pools, selectively targeting rapidly proliferating cells in abnormal skin, leading to inflammation, erosion, and lesion resolution. Its targeted cytotoxicity makes it a promising choice in dermato-oncology, effectively treating malignant and non-malignant skin conditions while minimizing harm to healthy skin cells [67,68,69,70,71]. Imiquimod acts as a Toll-like receptor 7 agonist and serves as a topical immunostimulatory agent. Treatment with imiquimod has achieved remission rates exceeding 70% in patients with localized cSCC, although surgery is still considered more effective overall [72]. However, imiquimod has not been shown to be effective in treating or preventing cSCC in immunocompetent patients [73]. Recent advances in topical immune-modulating treatments include the utilization of thymic stromal lymphopoietin (TSLP). Known for its strong antitumor properties in skin with compromised barriers, TSLP has shown considerable promise in enhancing skin cancer therapies [74]. It can be activated in the skin by calcipotriol (calcipotriene), a topical medication approved by the FDA for treating psoriasis [75]. A recent randomized, double-blind clinical trial with immunocompetent patients found that combining calcipotriol with 5-FU not only eradicated actinic keratoses (precursor lesions of cSCC) but also triggered a robust immune response involving CD4+ and CD8+ tissue-resident memory cells targeting premalignant epidermal clones. Three years later, the trial revealed that significantly fewer individuals in the calcipotriol plus 5-FU treatment group developed new cSCC cases compared to those in the control group [76,77].

3.2.2. Targeted Therapy

In the past, advanced cSCC was predominantly treated through targeted therapeutic strategies, particularly emphasizing the utilization of EGFR inhibitors [78,79]. However, with the advent of immunotherapy, the role of these agents has become less prominent. Among the most promising therapeutic agents are cetuximab and panitumumab.
The administration of cetuximab, whether independently or in combination with radiotherapy or platinum-based agents, has demonstrated clinical effectiveness in managing advanced cSCC. Phase II trials have investigated EGFR inhibitors, such as cetuximab, in patients with metastatic or locally advanced cSCC, displaying variable efficacy [80,81]. The objective response rate (ORR) ranged from 28% to 42%. Conversely, when used as a standalone treatment for cSCC, panitumumab has shown notable safety and efficacy profiles. Cetuximab is often utilized as a secondary-line option following primary or acquired resistance to a PD1 therapy, while panitumumab awaits formal approval for the treatment of cSCC despite demonstrating efficacy [82]. Genetic studies show a low prevalence of mutations in RAS, BRAF, and EGFR in cSCC, indicating that pre-screening before cetuximab therapy may not be necessary [83]. However, caution is recommended due to the potential for adverse events associated with cetuximab and panitumumab therapies, including infections and bleeding related to EGFR-targeted therapies.

3.2.3. Systemic Immunotherapy

Systemic immunotherapy has revolutionized the treatment landscape for advanced or metastatic non-melanoma skin cancers like cSCC, especially for patients who are unsuitable candidates for surgery or radiation therapy. Checkpoint inhibitors such as cemiplimab and pembrolizumab have emerged as key players, targeting pathways like PD-1/PD-L1 and significantly improving patient outcomes. The advent of immunotherapy has transformed the management of advanced non-melanoma skin cancers, offering new hope for patients previously facing limited treatment options and poor outcomes.
Cemiplimab, a high-affinity monoclonal antibody against PD-1, has gained regulatory approval from both the United States Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for managing advanced cSCC and locally advanced BCC, administered intravenously every three weeks at a dosage of 350 mg per infusion [84,85,86,87]. Clinical trials, including the landmark EMPOWER-CSCC-1 trial (n = 78), have demonstrated efficacy across metastatic and locally advanced cSCC cohorts with an ORR of 47%, CR rate of 6.7%, and tolerability, irrespective of PD-L1 expression status [86,87,88,89]. In regard to its safety, cemiplimab exhibits a toxicity profile comparable to that of other PD-1/PD-L1 inhibitors. Grade 3 or higher immune-related adverse effects (irAEs) were reported in 10% of patients, with the most common being pneumonitis (3%), diarrhea (2%), and fatigue (2%) [88]. Real-world data reaffirm cemiplimab’s effectiveness and safety, establishing it as a frontline option for unresectable or metastatic cSCC [89,90,91,92,93]. Neoadjuvant cemiplimab therapy has shown promising outcomes, achieving pathological remission in a significant proportion of patients before surgical excision.
Pembrolizumab, a humanized PD-1 blocking antibody, has emerged as a significant therapeutic option in managing cSCC, particularly for cases resistant to conventional therapies [94]. Pembrolizumab received FDA approval as a first-line treatment for locally advanced unresectable or metastatic cSCC based on the KEYNOTE-629 trial (n = 105), in which the patients were administered intravenously every three weeks at the dosage of 200 mg for up to 35 cycles or until progression [95,96]. Pembrolizumab achieved an overall ORR of 40.3% and CR rate of 12.6% in all populations, with an ORR of 50% in the locally advanced cohort and an ORR of 35.2% in the recurrent/metastatic group. Other studies, such as the CARSKIN trial (n = 57), further supported pembrolizumab’s efficacy as a first-line agent for unresectable cSCC, reporting an ORR of 41% at week 15 and a CR rate of 21% [97]. In conclusion, pembrolizumab demonstrates both efficacy and tolerability in the treatment of cSCC, showing promise in recurrent or metastatic settings and as a first-line therapy. A total of 11.9% of patients experienced Grade 3–5 immune-related adverse events (irAEs), including severe skin reactions, immune colitis, and immune hepatitis [96]. Tragically, two fatalities occurred due to treatment-related adverse events: one from cranial nerve neuropathy and another from immune-mediated colitis.

3.3. New Treatment Modalities

In addition to cetuximab and panitumumab, other EGFR inhibitors were explored in phase II studies, such as gefitinib (n = 40), erlotinib (n = 29), and lapatinib (n = 10). Gefitinib treatment of patients with recurrent and metastatic cSCC was conducted in a phase II study (NCT00054691) at The University of Texas M. D. Anderson Cancer Center. The treatment regimen consisted of administering gefitinib orally at a dose of 250 mg per day until either disease progression or the onset of intolerable side effects, showing an ORR of 16% [98]. In a phase 2 trial (NCT01198028), the efficacy of erlotinib was evaluated in patients with recurrent or metastatic cSCC who were unsuitable for curative treatment [99]. Thirty-nine patients received 150 mg of erlotinib daily. The overall response rate was 10%, and no unexpected toxicities were observed. A phase 2 clinical trial (NCT0166431) was conducted to investigate the impact of lapatinib on cutaneous squamous cell carcinoma (cSCC) and precursor lesions. Ten male patients received neoadjuvant lapatinib therapy, demonstrating a reduction in cSCC size in two patients and a 30% decrease in precursor lesions after 56 days [100]. These findings suggest potential for further research, particularly in high-risk patients.
The involvement of the mTOR pathway in cSCC and other NMSCs has attracted significant interest as well [101]. mTOR inhibitors have demonstrated potential as efficacious treatments, particularly in solid-organ transplant recipients. Topical rapamycin treatment has shown efficacy in conditions such as Paget’s disease and extramammary Paget’s disease (EMPD) [102,103,104]. In conclusion, while EGFR inhibitors remain pivotal, targeted therapies and mTOR inhibitors offer promising alternatives.
Two promising new PD-1 inhibitors effective in treating advanced and metastatic cSCC are nivolumab and cosibelimab. Nivolumab has also been investigated in the first-line treatment of advanced CSCC and in the second-line therapeutic option for head and neck cSCC in China [105]. A phase II investigation (NCT03834233) in which the patients were administered intravenously every two weeks at a dosage of 3 mg/kg for up to 12 months revealed encouraging results, with an ORR of 54.5% at 24 weeks for advanced cSCC, albeit accompanied by notable treatment-related AEs affecting 21% of patients (n = 24) [106]. Cosibelimab was studied in a phase I trial (NCT03212404) in patients with metastatic cSCC (n = 10) [107]. Administered intravenously at 800 mg every two weeks, it yielded an ORR of 47.4% among participants, suggesting potential as a therapy for metastatic cSCC with a tolerable safety profile. The most common adverse events included fatigue, rash, and anemia, with manageable immune-related adverse events reported in 23.1% of participants.
In addition to checkpoint inhibitors, alternative immunotherapeutic approaches such as intralesional injections of talimogene laherparepvec (T-VEC), an HSV-1 oncolytic virus FDA-approved for the local treatment of unresectable recurrent melanoma, are being investigated (NCT03714828). Clinical trials are exploring combination therapies to enhance response rates and improve patient prognosis [108,109]. In this pilot study, all seven participants achieved impressive ORR and CR values of 100%, with mild adverse effects. These findings highlight TVEC’s potential as a treatment option for unresectable cSCC (Table 1).

4. Basal Cell Carcinoma

4.1. Clinical Features and Staging of BCC

BCC is the most common form of human skin cancer arising from the basal layer of the epidermis [110]. It is more commonly observed in sun-exposed areas of the skin, including the face, neck, and trunk [111,112,113,114]. BCCs are typically small or intermediate-size lesions that progress slowly and display benign clinical prognosis. In some cases, local tissue infiltration and ulceration can occur, although BCC is rarely associated with the onset of metastasis [115]. Metastatic BCC and locally advanced BCC are distinguished by marked invasiveness, rapid growth rate, and tendency to recur [116,117]. These types of BCC often result in a poor prognosis [118]. The clinical features of BCC are diverse, and several variants have been described. However, BCC is typically classified into three categories, superficial, nodular, and infiltrative, with the last one being the least common and characterized by superficial blood vessels and local tissue invasion [110,119].
According to the NCCN, BCC staging is based on the likelihood of recurrence, which varies depending on the location of the tumor. Thus, low-risk, high-risk, and regional or distant metastasis are categorized into three categories. Low-risk BCCs are minor and superficial with clear and defined edges, while high-risk BCCs measure at least 2 cm in width and have returned despite therapy in the past. If a lesion has spread to a remote site, it is classified as regional or distant metastatic BCC. A novel classification released by the European Association of Dermato-Oncology (EADO) that considers factors including size, location, boundary definition, previous treatments, and related recurrences divided BCCs into two categories: easy-to-treat BCC and difficult-to-treat BCC [120,121,122]. Easy-to-treat BCC accounts for about 95% of BCCs with low recurrence risk. In comparison, difficult-to-treat BCC includes locally advanced and metastatic BCC, which have an estimated incidence of 0.8% and 0.0028–0.55%, respectively [123,124]. This classification is essential for determining the most appropriate treatment approach [114].

4.2. Current Available Therapies

Surgery is the primary treatment modality for easy-to-treat BCCs, which also offers the opportunity to perform a deeper examination of the safety margins and histological characteristics of the tumor [120,125,126]. MCS should be instead utilized in patients with high-risk BCC. However, it should be noted that surgery is associated with a high risk of relapse. Radiotherapy has been identified as a potential treatment alternative for patients who are not eligible for surgical intervention, with a comparable recurrence rate to that of surgery [127]. When surgery and radiotherapy are unsuitable or contraindicated for the patient, second-line treatment options may include topical treatment, targeted Hh inhibitors, or PD-1 inhibitor immunotherapy.
Figure 2 illustrates the algorithm for the treatment of BCC.

4.2.1. Topical Treatment

Topical treatments are indicated as an option in low-risk superficial BCCs [128]. Topical agents exhibit fewer side effects, are more cost-effective, and have lower cosmetic implications, especially in patients where multiple BCC lesions necessitate multiple excisions [129,130,131,132]. Imiquimod 5% and 5-fluorouracil 5% cream are currently FDA-authorized for treating small superficial BCCs [133,134,135]. Imiquimod is an immune response modifier that stimulates the release of inflammatory immunomodulatory cytokines, leading to apoptosis of BCC [136]. The treatment is indicated for use in immunocompetent adults and requires the application of the cream five times per week for six weeks. Fluorouracil is a chemotherapeutic agent that acts as a pyrimidine antimetabolite, interfering with DNA synthesis and inhibiting the proliferation of BCC cells [137,138]. It must be applied twice daily for two to four weeks [135]. The most common adverse effects are localized skin irritations, including burning and itching. The immune-modulating effect of imiquimod may be more effective in preventing superficial BCC recurrence [139,140]. The concerns regarding the utilization of topical treatment for BCC are related to the difficulties of self-application by patients, the potential adverse reactions, and the restricted application limited to low-grade superficial lesions.

4.2.2. Hedgehog Inhibitors

Aberrant activation of the Hh pathway and the subsequent loss of SMO receptor inhibition is a hallmark of basal cell carcinoma [141]. The loss of SMO inhibition induces a cascade activation that promotes tumor cell division and proliferation [142,143]. Identifying the role of Hh pathway mutations in BCC pathogenesis pioneered the development of Hh inhibitor treatment. Two oral Hh inhibitors, vismodegib and sonidegib, have been FDA-approved for patients with locally advanced BCC and metastatic BCC [144]. These drugs are semi-synthetic derivatives of natural alkaloid cyclopamine and target SMO protein [145]. Oral administration is convenient and preferred by patients due to non-invasiveness. However, adverse side effects, including muscle spasms and alopecia, may be alleviated with topical therapy [146,147]. Although Hh inhibitors are an effective alternative for treating advanced BCC, drug resistance is one of the significant concerns, and known SMO mutations can confer functional resistance to vismodegib and sonidegib [148,149]. Vismodegib is approved worldwide for treating adult patients with locally advanced BCC or metastatic BCC who are not candidates for surgery or radiotherapy [150,151]. It was the first authorized Hh inhibitor following the success of the phase I ERIVANCE trial in which patients with locally advanced BCC (n = 15) or metastatic BCC (n = 18) received 150 mg daily. The ORR was 43% and 30%, respectively, and the results were in line with subsequent studies like the STEVIE trial, which enrolled 1215 subjects with an ORR of 69% in the locally advanced BCC group and 37% in patients with metastatic BCC [152]. However, the common adverse effects such as muscle spasms, alopecia, fatigue, and nausea have a high incidence and often result in treatment discontinuation [153]. In addition, the rate of intrinsic or acquired resistance to vismodegib is as high as 20% within the first year of treatment [154]. At a daily dose of 200 mg, sonidegib is the second FDA-approved drug for treating unresectable locally advanced BCC [155,156]. In fact, in the BOLT trial of patients with locally advanced BCC and metastatic BCC receiving sonidegib at 800 mg and 200 mg daily, tumor response was independent of sonidegib dose in this range, suggesting 200 mg/day instead of 800 mg/day [157]. The final report at 42 months showed that ORRs were 56% for locally advanced BCC and 8% for metastatic BCC in the 200 mg group and 46.1% and 17%, respectively, in the 800 mg cohort [158]. The most common adverse effects were similar to those reported with vismodegib but also included raised creatinine and lipase concentration [157].

4.2.3. Systemic Immunotherapy

BCC cells express PD-L1, which, through the binding of the PD-1, can lead to the inhibition of T cell function [159,160]. Cemiplimab is an anti-PD-1 monoclonal antibody that enhances antitumor responses. It has been FDA-approved for treating locally advanced BCC and metastatic BCC in patients with progressive disease, resistance or intolerance to Hh inhibitors [161]. In a study involving 84 locally advanced BCC patients, cemiplimab was administered intravenously at a dosage of 350 mg every three weeks for up to 93 weeks. The ORR was 31%, including 6% CR and 25% PR [162]. However, 97% of participants had adverse effects, most commonly fatigue and itching, but colitis and adrenal insufficiency were observed [162].

4.3. New Treatment Modalities

New clinical trials are underway for BCC, some of which have reported successful preliminary results. Patidegib is a novel SMO inhibitor under development for topical and oral administration. In a study involving 36 subjects with nodular BCC, patients were treated topically once or twice daily with patidegib 2% or 4% gel for a total of 12 weeks, with patidegib 2% gel showing a greater efficacy and lower adverse effects (NCT02828111). Meanwhile, oral administration of patidegib at various doses (20 to 210 mg/day) was associated with the most commonly reported adverse effects of muscle spasms, fatigue, nausea, and hair loss in a phase I study involving 94 patients with advanced BCC [163]. Other studies are ongoing, and promising outcomes have been evaluated, evaluating the efficacy of oral administration of second-generation Hh inhibitors such as taladegib and itraconazole (NCT03972748) [164,165,166]. Topical itraconazole treatment is assessed in two phase I trials (NCT02735356). Regarding immunotherapy, there has also been progress in clinical trials on PD-1 inhibitors. Recent studies have investigated the neoadjuvant use of oral pembrolizumab (NCT02690948) and nivolumab (NCT03521830) (Table 2).

5. Merkel Cell Carcinoma

5.1. Clinical Features and Staging of MCC

MCC is a rare and aggressive skin cancer with two main types: the virus-negative MCC (VN-MCC), driven by ultraviolet-induced mutations, and virus-positive MCC (VP-MCC), driven by Merkel cell polyomavirus (MPyV) [38,167]. Clinically, MCC presents as painless but rapidly expanding flesh-colored nodules, mainly in the skin or subcutaneous tissue [168]. Usually, MCC lesions are on sun-exposed skin, most commonly on the extremities, head, and neck. MCCs are often confused with other lesions like cysts or melanomas. Diagnosis requires biopsy due to nonspecific clinical features. However, 89% of patients with MCC show at least three of these features captured in the acronym AEIOU: asymptomatic, expanding rapidly, immunosuppression, older than age 50, and UV radiation [169]. Confirmation involves immunohistochemistry for neuroendocrine and epithelial markers such as cytokeratin 20. MCCs can metastasize without a primary lesion.
The 8th Edition AJCC staging system for MCC refines staging by distinguishing between clinical and pathological assessments using TNM criteria [170]. The majority of patients exhibit local (65%), nodal (26%), or distant (8%) disease. Survival outcomes are better for patients with pathological staging data. An extensive evaluation, encompassing physical examination and imaging, is vital for precise staging. If metastasis is not identified, patients may undergo a sentinel lymph node biopsy (SLNB) in conjunction with primary tumor surgery [171,172]. This approach facilitates the discussion of prognosis and treatment planning for patients with MCC.

5.2. Current Available Therapies

Surgical excision remains the primary treatment for resectable MCC. The standard procedure involves excising the primary tumor with 1 cm lateral margins, encompassing the underlying fascia, followed by a potential SLNB [173,174]. Adjuvant radiation therapy (50–55 Grays) is recommended post-surgery, also in patients with pathologically negative lymph nodes. For patients who are unsuitable candidates for surgery or have primary tumors in anatomical locations where excision would cause notable functional limitations, radiotherapy monotherapy serves as an alternative [175,176]. A positive SLNB without distant metastasis may result in complete lymph node dissection (CLND), radiation therapy, or both [174]. Treatment options encompass radiotherapy, immunotherapy, or chemotherapy in metastasis to additional organs.
Figure 3 illustrates the algorithm for the treatment of MCC.

Systemic Immunotherapy

The advent of checkpoint inhibitors targeting the PD-1/PD-L1 pathway has revolutionized the treatment of metastatic MCC, now recommended as the first-line therapy by global guidelines. Avelumab, the first systemic therapy for MCC, proved effective in both second-line (n = 88) and first-line (n = 116) treatments in the JAVELIN Merkel 200 trial [7,177]. Administered intravenously every two weeks at a dosage of 10 mg/kg until treatment progression, it achieved an ORR of 33% and CR of 11% post-chemotherapy failure. In first-line treatment, a 40% ORR was observed. While PD-L1 positivity correlated with better responses, it was not a requisite for the treatment. Although not approved for the treatment of locally advanced disease alone, the off-label use of this treatment is being explored [178,179]. Pembrolizumab gained FDA approval in 2018 for recurrent locally advanced or metastatic MCC. In Keynote 017 (n = 50), in which the patients were administered intravenously every three weeks at a 2 mg/kg dosage for up to 2 years, the ORR was 58%, including 24% demonstrating an imaging CR [180,181]. Retifanlimab, another PD-1 receptor monoclonal antibody, was investigated in the POD1UM-201 phase II trial (NCT03599713), in which 87 treatment-naïve patients received 500 mg IV every four weeks for up to 2 years, with an ORR of 46% and CR of 12%. Subsequently, the FDA approved retifanlimab for adult patients with metastatic or recurrent locally advanced MC.

5.3. New Treatment Modalities

Targeted therapies represent a promising avenue for patients with advanced MCC who are ineligible for or unresponsive to immune checkpoint inhibitors. Merkel cell carcinoma exhibits increased angiogenesis driven by high VEGF-A expression. Trials with tyrosine kinase inhibitors (TKIs), such as pazopanib and cabozantinib, targeting VEGFR and other pathways, showed modest efficacy [182]. The use of TKIs in combination with immunotherapies is currently being investigated to improve outcomes (NCT04869137). YM155 and ABT-263, small-molecule inhibitors of antiapoptotic protein surviving and apoptosis regulator BCL-2 family proteins, have shown efficacy in inducing cell death in MCC cell lines and xenografts [183,184]. VN-MCC is frequently associated with p53 mutations, whereas VP-MCC is typically characterized by wild-type p53 [185,186]. KRT-232 (navtemadlin), an MDM2 inhibitor, demonstrated efficacy in phase 1b/2 trials (NCT03787602) for p53 wild-type MCC patients, particularly those resistant to anti-PD1/PD-L1 therapy. Despite these promising results, drug development for MCC was discontinued.
Merkel cell carcinoma expresses somatostatin receptors (SSTRs), suggesting potential avenues for treatment with somatostatin analogs such as octreotide and lanreotide [187,188]. Although the effectiveness of these treatments in MCC is limited, ongoing investigations aim to enhance outcomes by developing higher-affinity analogs and peptide receptor radionuclide therapy.
The PI3K/AKT/mTOR signaling pathway is crucial in promoting tumor cell proliferation and survival, including in the MCC [189]. MCC often exhibits elevated levels of phosphorylated AKT and occasional mutations in the PIK3CA gene, indicating pathway activation [190,191]. Inhibition of mTOR has shown promise in inducing cell death and suppressing MCC growth in preclinical models [192,193]. Despite setbacks in clinical trials targeting both mTOR complexes, recent research continues to highlight the importance of this pathway in MCC, with upregulated gene expression linked to poor outcomes [194]. Clinical responses to PI3K and AKT inhibitors underscore the therapeutic potential of targeting this pathway. There has only been a single reported case of a patient with advanced MCC carrying a known PI3K mutation successfully treated with idelalisib, a PI3K-δ inhibitor, resulting in a rapid and complete remission [195]. A clinical trial is investigating the safety and efficacy of mTOR inhibition in patients with MCC (NCT02514824). Studies have demonstrated the effectiveness of copanlisib in inhibiting tumor growth, while combination therapies with alpelisib and the BCL-2 inhibitor navitoclax show synergistic effects [196,197]. Additionally, artesunate, known for its anti-malarial properties, exhibits cytotoxic activity in MCC, particularly in MCPyV-positive cases, suggesting a novel therapeutic avenue for further exploration [198,199].
A recent preclinical study highlighted MUC1-C as a potential therapeutic target in MCC [175,200]. This study revealed that MUC1-C interacts with MYCL, promoting MCC progression and cell survival in both VP- and VN-MCC cells [201]. This finding underscores the significance of targeting MUC1-C in MCC treatment, offering a potential avenue for pharmacological intervention to impede disease progression.
In addition to the previous PD-1/PD-L1 inhibitors, nivolumab has been investigated in treating advanced MCC, achieving an ORR of 68% (CheckMate 358 phase I/II trial, NCT02488759). Combining nivolumab with ipilimumab, targeting cytotoxic T-lymphocyte associated antigen-4 (CTLA-4), enhances antitumor responses, offering potential for patients unresponsive to PD-1 blockade [202,203]. CTLA-4 is an immune checkpoint protein able to repress the function of activated T cells binding CD80 or CD86 on their surface [204]. Despite the challenges that remain, immune checkpoint inhibitors represent a pivotal advancement in the management of MCC, offering hope for prolonged survival and enhanced quality of life among patients with this aggressive malignancy.
Other potential treatment avenues under investigation include immunostimulatory agents such as MCPyV vaccination and talimogene laherparepvec (T-VEC). In situ vaccination with plasmid DNA has been demonstrated to be an effective treatment for advanced MCC patients who have shown resistance to PD-1/PD-L1 inhibitors. At the same time, off-label use of T-VEC showed promise in treating recurrent MCC, with reported partial or complete responses in several patients [205,206,207,208,209]. Clinical trials explore its potential in MCC and other non-melanoma cancers (NCT03458117, NCT02819843, and NCT02978625). These treatments may offer alternative options, particularly for patients unsuited for immunotherapy (Table 3).

6. Future Perspectives

Recent advancements in understanding the pathogenesis of cSCC, BCC, and MCC have significantly advanced therapeutic options, enhancing patients’ survival and quality of life. Crucial findings have highlighted the role of immune suppression in cSCC, the Hedgehog pathway’s critical involvement in BCC and its targeted inhibition’s success, and the MPyV’s influence in the development of VP- MCC. Systemic immunotherapies targeting PD-1 and PD-L1 have transformed the management of aggressive and metastatic NMSC. However, the etiological complexity of NMSC, involving both genetic and environmental factors, prevents a singular treatment approach. Furthermore, the incidence of NMSC has grown over time and this is correlated with growing treatment costs. Of the NMSC treatments that have been addressed in this review, topical therapy is more cost-effective than other non-surgical therapies since it requires fewer in-office procedures and reduces surgical risks such as scarring and infection [210]. However, as with surgery, the cost of treatment is influenced by the size of the tumor and the delay in diagnosis. It is evident that the future of NMSC management will increasingly rely on successful primary and secondary prevention strategies. Well-defined risk factors, such as UV exposure and immune suppression, provide unique opportunities for preventive strategies not typically available for other cancers. Recent studies have also highlighted the dynamic interactions within the tumor microenvironment, offering new perspectives on NMSC management, including therapies for metastatic cSCC and extending research into MCC and BCC. Future treatments may need to address the continuously evolving nature of cancer cells and the intricate interplay of genetic, immunological, and stromal factors. Adopting a holistic approach could revolutionize treatment strategies by focusing not only on eliminating cancer cells but also on altering the broader tumor environment to inhibit cancer progression.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare the following financial interests, which may be considered potential competing interests: AM holds equity in DermBiont, Inc., a company conducting clinical trials to assess a novel treatment for BCC and SCC. SS, FB, KT, and SEW report no conflicts of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  2. Larese Filon, F.; Buric, M.; Fluehler, C. UV exposure, preventive habits, risk perception, and occupation in NMSC patients: A case-control study in Trieste (NE Italy). Photodermatol. Photoimmunol. Photomed. 2019, 35, 24–30. [Google Scholar] [CrossRef] [PubMed]
  3. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed]
  4. Tanese, K.; Nakamura, Y.; Hirai, I.; Funakoshi, T. Updates on the Systemic Treatment of Advanced Non-melanoma Skin Cancer. Front. Med. 2019, 6, 160. [Google Scholar] [CrossRef]
  5. Wysong, A.; Aasi, S.Z.; Tang, J.Y. Update on metastatic basal cell carcinoma: A summary of published cases from 1981 through 2011. JAMA Dermatol. 2013, 149, 615–616. [Google Scholar] [CrossRef]
  6. Zhu, G.A.; Lynn Su Chang, A. Overall and progression-free survival of stage 4 cutaneous squamous cell carcinoma at a single large referral center. J. Am. Acad. Dermatol. 2015, 73, 165–166. [Google Scholar] [CrossRef] [PubMed]
  7. D’Angelo, S.P.; Bhatia, S.; Brohl, A.S.; Hamid, O.; Mehnert, J.M.; Terheyden, P.; Shih, K.C.; Brownell, I.; Lebbe, C.; Lewis, K.D.; et al. Avelumab in patients with previously treated metastatic Merkel cell carcinoma: Long-term data and biomarker analyses from the single-arm phase 2 JAVELIN Merkel 200 trial. J. Immunother. Cancer 2020, 8, e000674. [Google Scholar] [CrossRef]
  8. Lv, R.; Sun, Q. A Network Meta-Analysis of Non-Melanoma Skin Cancer (NMSC) Treatments: Efficacy and Safety Assessment. J. Cell. Biochem. 2017, 118, 3686–3695. [Google Scholar] [CrossRef] [PubMed]
  9. Bhandari, P.R.; Pai, V.V. Novel medical strategies combating nonmelanoma skin cancer. Indian J. Dermatol. 2014, 59, 531–546. [Google Scholar] [CrossRef]
  10. Thompson, A.K.; Kelley, B.F.; Prokop, L.J.; Murad, M.H.; Baum, C.L. Risk Factors for Cutaneous Squamous Cell Carcinoma Recurrence, Metastasis, and Disease-Specific Death: A Systematic Review and Meta-analysis. JAMA Dermatol. 2016, 152, 419–428. [Google Scholar] [CrossRef]
  11. Collins, L.; Quinn, A.; Stasko, T. Skin Cancer and Immunosuppression. Dermatol. Clin. 2019, 37, 83–94. [Google Scholar] [CrossRef]
  12. Staples, M.P.; Elwood, M.; Burton, R.C.; Williams, J.L.; Marks, R.; Giles, G.G. Non-melanoma skin cancer in Australia: The 2002 national survey and trends since 1985. Med. J. Aust. 2006, 184, 6–10. [Google Scholar] [CrossRef]
  13. Subramaniam, P.; Olsen, C.M.; Thompson, B.S.; Whiteman, D.C.; Neale, R.E.; Sun, Q.S.; Health Study, I. Anatomical Distributions of Basal Cell Carcinoma and Squamous Cell Carcinoma in a Population-Based Study in Queensland, Australia. JAMA Dermatol. 2017, 153, 175–182. [Google Scholar] [CrossRef]
  14. Qureshi, A.A.; Laden, F.; Colditz, G.A.; Hunter, D.J. Geographic variation and risk of skin cancer in US women. Differences between melanoma, squamous cell carcinoma, and basal cell carcinoma. Arch. Intern. Med. 2008, 168, 501–507. [Google Scholar] [CrossRef] [PubMed]
  15. Brenner, M.; Hearing, V.J. The protective role of melanin against UV damage in human skin. Photochem. Photobiol. 2008, 84, 539–549. [Google Scholar] [CrossRef] [PubMed]
  16. Brand, R.M.; Wipf, P.; Durham, A.; Epperly, M.W.; Greenberger, J.S.; Falo, L.D., Jr. Targeting Mitochondrial Oxidative Stress to Mitigate UV-Induced Skin Damage. Front. Pharmacol. 2018, 9, 920. [Google Scholar] [CrossRef]
  17. Drobetsky, E.A.; Grosovsky, A.J.; Glickman, B.W. The specificity of UV-induced mutations at an endogenous locus in mammalian cells. Proc. Natl. Acad. Sci. USA 1987, 84, 9103–9107. [Google Scholar] [CrossRef]
  18. Dachani, S.R.; Kaleem, M.; Mujtaba, M.A.; Mahajan, N.; Ali, S.A.; Almutairy, A.F.; Mahmood, D.; Anwer, M.K.; Ali, M.D.; Kumar, S. A Comprehensive Review of Various Therapeutic Strategies for the Management of Skin Cancer. ACS Omega 2024, 9, 10030–10048. [Google Scholar] [CrossRef] [PubMed]
  19. Lai, M.; Pampena, R.; Cornacchia, L.; Odorici, G.; Piccerillo, A.; Pellacani, G.; Peris, K.; Longo, C. Cutaneous squamous cell carcinoma in patients with chronic lymphocytic leukemia: A systematic review of the literature. Int. J. Dermatol. 2022, 61, 548–557. [Google Scholar] [CrossRef]
  20. Yeung, H.; Balakrishnan, V.; Luk, K.M.H.; Chen, S.C. Risk of Skin Cancers in Older Persons Living with HIV: A Systematic Review. J. Assoc. Nurses AIDS Care 2019, 30, 80–86. [Google Scholar] [CrossRef]
  21. Lindelof, B.; Sigurgeirsson, B.; Gabel, H.; Stern, R.S. Incidence of skin cancer in 5356 patients following organ transplantation. Br. J. Dermatol. 2000, 143, 513–519. [Google Scholar] [PubMed]
  22. Tessari, G.; Girolomoni, G. Nonmelanoma skin cancer in solid organ transplant recipients: Update on epidemiology, risk factors, and management. Dermatol. Surg. 2012, 38, 1622–1630. [Google Scholar] [CrossRef] [PubMed]
  23. Matinfar, M.; Shahidi, S.; Feizi, A. Incidence of nonmelanoma skin cancer in renal transplant recipients: A systematic review and meta-analysis. J. Res. Med. Sci. 2018, 23, 14. [Google Scholar] [CrossRef] [PubMed]
  24. Stenz, N.A.; Stampf, S.; Arnold, A.W.; Cozzio, A.; Dickenmann, M.; Gaide, O.; Harms, M.; Hunger, R.E.; Laffitte, E.; Muhlstadt, M.; et al. Skin Cancer Development in Solid Organ Transplant Recipients in Switzerland (Swiss Transplant Cohort Study). Dermatology 2021, 237, 970–980. [Google Scholar] [CrossRef] [PubMed]
  25. Chalmers, Z.R.; Connelly, C.F.; Fabrizio, D.; Gay, L.; Ali, S.M.; Ennis, R.; Schrock, A.; Campbell, B.; Shlien, A.; Chmielecki, J.; et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017, 9, 34. [Google Scholar] [CrossRef] [PubMed]
  26. Knepper, T.C.; Montesion, M.; Russell, J.S.; Sokol, E.S.; Frampton, G.M.; Miller, V.A.; Albacker, L.A.; McLeod, H.L.; Eroglu, Z.; Khushalani, N.I.; et al. The Genomic Landscape of Merkel Cell Carcinoma and Clinicogenomic Biomarkers of Response to Immune Checkpoint Inhibitor Therapy. Clin. Cancer Res. 2019, 25, 5961–5971. [Google Scholar] [CrossRef] [PubMed]
  27. Winge, M.C.G.; Kellman, L.N.; Guo, K.; Tang, J.Y.; Swetter, S.M.; Aasi, S.Z.; Sarin, K.Y.; Chang, A.L.S.; Khavari, P.A. Advances in cutaneous squamous cell carcinoma. Nat. Rev. Cancer 2023, 23, 430–449. [Google Scholar] [CrossRef] [PubMed]
  28. Sarin, K.Y.; Lin, Y.; Daneshjou, R.; Ziyatdinov, A.; Thorleifsson, G.; Rubin, A.; Pardo, L.M.; Wu, W.; Khavari, P.A.; Uitterlinden, A.; et al. Genome-wide meta-analysis identifies eight new susceptibility loci for cutaneous squamous cell carcinoma. Nat. Commun. 2020, 11, 820. [Google Scholar] [CrossRef] [PubMed]
  29. Tate, J.G.; Bamford, S.; Jubb, H.C.; Sondka, Z.; Beare, D.M.; Bindal, N.; Boutselakis, H.; Cole, C.G.; Creatore, C.; Dawson, E.; et al. COSMIC: The Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res. 2019, 47, D941–D947. [Google Scholar] [CrossRef]
  30. Piipponen, M.; Riihila, P.; Nissinen, L.; Kahari, V.M. The Role of p53 in Progression of Cutaneous Squamous Cell Carcinoma. Cancers 2021, 13, 4507. [Google Scholar] [CrossRef]
  31. Campos, M.A.; Macedo, S.; Fernandes, M.S.; Pestana, A.; Pardal, J.; Batista, R.; Vinagre, J.; Sanches, A.; Baptista, A.; Lopes, J.M.; et al. Prognostic Significance of RAS Mutations and P53 Expression in Cutaneous Squamous Cell Carcinomas. Genes 2020, 11, 751. [Google Scholar] [CrossRef] [PubMed]
  32. Wang, N.J.; Sanborn, Z.; Arnett, K.L.; Bayston, L.J.; Liao, W.; Proby, C.M.; Leigh, I.M.; Collisson, E.A.; Gordon, P.B.; Jakkula, L.; et al. Loss-of-function mutations in Notch receptors in cutaneous and lung squamous cell carcinoma. Proc. Natl. Acad. Sci. USA 2011, 108, 17761–17766. [Google Scholar] [CrossRef] [PubMed]
  33. South, A.P.; Purdie, K.J.; Watt, S.A.; Haldenby, S.; den Breems, N.; Dimon, M.; Arron, S.T.; Kluk, M.J.; Aster, J.C.; McHugh, A.; et al. NOTCH1 mutations occur early during cutaneous squamous cell carcinogenesis. J. Investig. Dermatol. 2014, 134, 2630–2638. [Google Scholar] [CrossRef] [PubMed]
  34. de Lima, P.O.; Joseph, S.; Panizza, B.; Simpson, F. Epidermal Growth Factor Receptor’s Function in Cutaneous Squamous Cell Carcinoma and Its Role as a Therapeutic Target in the Age of Immunotherapies. Curr. Treat. Opt. Oncol. 2020, 21, 9. [Google Scholar] [CrossRef] [PubMed]
  35. Hernandez, L.E.; Mohsin, N.; Levin, N.; Dreyfuss, I.; Frech, F.; Nouri, K. Basal cell carcinoma: An updated review of pathogenesis and treatment options. Dermatol. Ther. 2022, 35, e15501. [Google Scholar] [CrossRef] [PubMed]
  36. 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] [PubMed]
  37. Chmiel, P.; Klosinska, M.; Forma, A.; Pelc, Z.; Geca, K.; Skorzewska, M. Novel Approaches in Non-Melanoma Skin Cancers—A Focus on Hedgehog Pathway in Basal Cell Carcinoma (BCC). Cells 2022, 11, 3210. [Google Scholar] [CrossRef] [PubMed]
  38. Becker, J.C.; Stang, A.; DeCaprio, J.A.; Cerroni, L.; Lebbe, C.; Veness, M.; Nghiem, P. Merkel cell carcinoma. Nat. Rev. Dis. Primers 2017, 3, 17077. [Google Scholar] [CrossRef] [PubMed]
  39. Wijaya, W.A.; Liu, Y.; Qing, Y.; Li, Z. Prevalence of Merkel Cell Polyomavirus in Normal and Lesional Skin: A Systematic Review and Meta-Analysis. Front. Oncol. 2022, 12, 868781. [Google Scholar] [CrossRef]
  40. Garneski, K.M.; Warcola, A.H.; Feng, Q.; Kiviat, N.B.; Leonard, J.H.; Nghiem, P. Merkel cell polyomavirus is more frequently present in North American than Australian Merkel cell carcinoma tumors. J. Investig. Dermatol. 2009, 129, 246–248. [Google Scholar] [CrossRef]
  41. Woodhouse, B.; Robb, T.J.; Hearn, J.I.; Houseman, P.S.; Hayward, G.; Miller, R.; Restall, A.P.; Findlay, M.; Lawrence, B.; Print, C.G.; et al. Merkel cell polyomavirus is uncommon in New Zealand Merkel cell carcinomas. Br. J. Dermatol. 2018, 179, 1197–1198. [Google Scholar] [CrossRef] [PubMed]
  42. Goh, G.; Walradt, T.; Markarov, V.; Blom, A.; Riaz, N.; Doumani, R.; Stafstrom, K.; Moshiri, A.; Yelistratova, L.; Levinsohn, J.; et al. Mutational landscape of MCPyV-positive and MCPyV-negative Merkel cell carcinomas with implications for immunotherapy. Oncotarget 2016, 7, 3403–3415. [Google Scholar] [CrossRef] [PubMed]
  43. Shuda, M.; Feng, H.; Kwun, H.J.; Rosen, S.T.; Gjoerup, O.; Moore, P.S.; Chang, Y. T antigen mutations are a human tumor-specific signature for Merkel cell polyomavirus. Proc. Natl. Acad. Sci. USA 2008, 105, 16272–16277. [Google Scholar] [CrossRef] [PubMed]
  44. Hesbacher, S.; Pfitzer, L.; Wiedorfer, K.; Angermeyer, S.; Borst, A.; Haferkamp, S.; Scholz, C.J.; Wobser, M.; Schrama, D.; Houben, R. RB1 is the crucial target of the Merkel cell polyomavirus Large T antigen in Merkel cell carcinoma cells. Oncotarget 2016, 7, 32956–32968. [Google Scholar] [CrossRef]
  45. Houben, R.; Adam, C.; Baeurle, A.; Hesbacher, S.; Grimm, J.; Angermeyer, S.; Henzel, K.; Hauser, S.; Elling, R.; Brocker, E.B.; et al. An intact retinoblastoma protein-binding site in Merkel cell polyomavirus large T antigen is required for promoting growth of Merkel cell carcinoma cells. Int. J. Cancer 2012, 130, 847–856. [Google Scholar] [CrossRef] [PubMed]
  46. Fania, L.; Didona, D.; Di Pietro, F.R.; Verkhovskaia, S.; Morese, R.; Paolino, G.; Donati, M.; Ricci, F.; Coco, V.; Ricci, F.; et al. Cutaneous Squamous Cell Carcinoma: From Pathophysiology to Novel Therapeutic Approaches. Biomedicines 2021, 9, 171. [Google Scholar] [CrossRef] [PubMed]
  47. Paolino, G.; Donati, M.; Didona, D.; Mercuri, S.R.; Cantisani, C. Histology of Non-Melanoma Skin Cancers: An Update. Biomedicines 2017, 5, 71. [Google Scholar] [CrossRef]
  48. Firnhaber, J.M. Basal Cell and Cutaneous Squamous Cell Carcinomas: Diagnosis and Treatment. Am. Fam. Physician 2020, 102, 339–346. [Google Scholar] [PubMed]
  49. Cassarino, D.S.; Derienzo, D.P.; Barr, R.J. Cutaneous squamous cell carcinoma: A comprehensive clinicopathologic classification. Part one. J. Cutan. Pathol. 2006, 33, 191–206. [Google Scholar] [CrossRef]
  50. Palaniappan, V.; Karthikeyan, K. Bowen’s Disease. Indian Dermatol. Online J. 2022, 13, 177–189. [Google Scholar] [CrossRef]
  51. Brancaccio, G.; Briatico, G.; Pellegrini, C.; Rocco, T.; Moscarella, E.; Fargnoli, M.C. Risk Factors and Diagnosis of Advanced Cutaneous Squamous Cell Carcinoma. Dermatol. Pract. Concept. 2021, 11, e2021166S. [Google Scholar] [CrossRef] [PubMed]
  52. Ebrahimi, A.; Gupta, R.; Luk, P.; Low, T.H.; McDowell, L.; Magarey, M.J.R.; Smith, P.N.; Perriman, D.M.; Schulte, K.M.; Veness, M.; et al. Number of nodal metastases and the American Joint Committee on cancer staging of head and neck cutaneous squamous cell carcinoma: A multicenter study. Oral Oncol. 2020, 111, 104855. [Google Scholar] [CrossRef] [PubMed]
  53. Xu, M.J.; Lazar, A.A.; Garsa, A.A.; Arron, S.T.; Ryan, W.R.; El-Sayed, I.H.; George, J.R.; Algazi, A.P.; Heaton, C.M.; Ha, P.K.; et al. Major prognostic factors for recurrence and survival independent of the American Joint Committee on Cancer eighth edition staging system in patients with cutaneous squamous cell carcinoma treated with multimodality therapy. Head Neck 2018, 40, 1406–1414. [Google Scholar] [CrossRef] [PubMed]
  54. Karia, P.S.; Jambusaria-Pahlajani, A.; Harrington, D.P.; Murphy, G.F.; Qureshi, A.A.; Schmults, C.D. Evaluation of American Joint Committee on Cancer, International Union against Cancer, and Brigham and Women’s Hospital tumor staging for cutaneous squamous cell carcinoma. J. Clin. Oncol. 2014, 32, 327–334. [Google Scholar] [CrossRef] [PubMed]
  55. Work, G.; Invited, R.; Kim, J.Y.S.; Kozlow, J.H.; Mittal, B.; Moyer, J.; Olenecki, T.; Rodgers, P. Guidelines of care for the management of cutaneous squamous cell carcinoma. J. Am. Acad. Dermatol. 2018, 78, 560–578. [Google Scholar] [CrossRef]
  56. Jambusaria-Pahlajani, A.; Kanetsky, P.A.; Karia, P.S.; Hwang, W.T.; Gelfand, J.M.; Whalen, F.M.; Elenitsas, R.; Xu, X.; Schmults, C.D. Evaluation of AJCC tumor staging for cutaneous squamous cell carcinoma and a proposed alternative tumor staging system. JAMA Dermatol. 2013, 149, 402–410. [Google Scholar] [CrossRef]
  57. Ruiz, E.S.; Karia, P.S.; Besaw, R.; Schmults, C.D. Performance of the American Joint Committee on Cancer Staging Manual, 8th Edition vs the Brigham and Women’s Hospital Tumor Classification System for Cutaneous Squamous Cell Carcinoma. JAMA Dermatol. 2019, 155, 819–825. [Google Scholar] [CrossRef]
  58. Kwiek, B.; Schwartz, R.A. Keratoacanthoma (KA): An update and review. J. Am. Acad. Dermatol. 2016, 74, 1220–1233. [Google Scholar] [CrossRef]
  59. Schmults, C.D.; Karia, P.S.; Carter, J.B.; Han, J.; Qureshi, A.A. Factors predictive of recurrence and death from cutaneous squamous cell carcinoma: A 10-year, single-institution cohort study. JAMA Dermatol. 2013, 149, 541–547. [Google Scholar] [CrossRef]
  60. Brougham, N.D.; Dennett, E.R.; Cameron, R.; Tan, S.T. The incidence of metastasis from cutaneous squamous cell carcinoma and the impact of its risk factors. J. Surg. Oncol. 2012, 106, 811–815. [Google Scholar] [CrossRef]
  61. Hunt, W.T.N.; Earp, E.; Brown, A.C.; Veitch, D.; Wernham, A.G.H. A review of Mohs micrographic surgery for skin cancer. Part 3: Squamous cell carcinoma. Clin. Exp. Dermatol. 2022, 47, 1765–1773. [Google Scholar] [CrossRef] [PubMed]
  62. Caccialanza, M.; Piccinno, R.; Moretti, D.; Rozza, M. Radiotherapy of carcinomas of the skin overlying the cartilage of the nose: Results in 405 lesions. Eur. J. Dermatol. 2003, 13, 462–465. [Google Scholar]
  63. Hernandez-Machin, B.; Borrego, L.; Gil-Garcia, M.; Hernandez, B.H. Office-based radiation therapy for cutaneous carcinoma: Evaluation of 710 treatments. Int. J. Dermatol. 2007, 46, 453–459. [Google Scholar] [CrossRef] [PubMed]
  64. Paasch, U.; Zidane, M.; Baron, J.M.; Bund, T.; Cappius, H.J.; Drosner, M.; Feise, K.; Fischer, T.; Gauglitz, G.; Gerber, P.A.; et al. S2k guideline: Laser therapy of the skin. J. Dtsch. Dermatol. Ges. 2022, 20, 1248–1267. [Google Scholar] [CrossRef] [PubMed]
  65. Lansbury, L.; Bath-Hextall, F.; Perkins, W.; Stanton, W.; Leonardi-Bee, J. Interventions for non-metastatic squamous cell carcinoma of the skin: Systematic review and pooled analysis of observational studies. BMJ 2013, 347, f6153. [Google Scholar] [CrossRef] [PubMed]
  66. Kwiatkowski, S.; Knap, B.; Przystupski, D.; Saczko, J.; Kedzierska, E.; Knap-Czop, K.; Kotlinska, J.; Michel, O.; Kotowski, K.; Kulbacka, J. Photodynamic therapy—Mechanisms, photosensitizers and combinations. Biomed. Pharmacother. 2018, 106, 1098–1107. [Google Scholar] [CrossRef] [PubMed]
  67. Prince, G.T.; Cameron, M.C.; Fathi, R.; Alkousakis, T. Topical 5-fluorouracil in dermatologic disease. Int. J. Dermatol. 2018, 57, 1259–1264. [Google Scholar] [CrossRef] [PubMed]
  68. Searle, T.; Al-Niaimi, F.; Ali, F.R. 5-Fluorouracil in Dermatology: The Diverse Uses Beyond Malignant and Premalignant Skin Disease. Dermatol. Surg. 2021, 47, e66–e70. [Google Scholar] [CrossRef]
  69. Ceilley, R.I. Mechanisms of action of topical 5-fluorouracil: Review and implications for the treatment of dermatological disorders. J. Dermatol. Treat. 2012, 23, 83–89. [Google Scholar] [CrossRef]
  70. Sethy, C.; Kundu, C.N. 5-Fluorouracil (5-FU) resistance and the new strategy to enhance the sensitivity against cancer: Implication of DNA repair inhibition. Biomed. Pharmacother. 2021, 137, 111285. [Google Scholar] [CrossRef]
  71. Metterle, L.; Nelson, C.; Patel, N. Intralesional 5-fluorouracil (FU) as a treatment for nonmelanoma skin cancer (NMSC): A review. J. Am. Acad. Dermatol. 2016, 74, 552–557. [Google Scholar] [CrossRef]
  72. Patel, G.K.; Goodwin, R.; Chawla, M.; Laidler, P.; Price, P.E.; Finlay, A.Y.; Motley, R.J. Imiquimod 5% cream monotherapy for cutaneous squamous cell carcinoma in situ (Bowen’s disease): A randomized, double-blind, placebo-controlled trial. J. Am. Acad. Dermatol. 2006, 54, 1025–1032. [Google Scholar] [CrossRef]
  73. Cheng, B.; Veerabagu, S.; Miller, C.J.; Sobanko, J.F.; Shin, T.M.; Higgins, H.W., 2nd; Giordano, C.N.; Etzkorn, J.R. A comparison of invasive squamous cell carcinoma greater than 1 year after treatment with 5-fluorouracil, imiquimod, or photodynamic therapy with aminolevulinic acid. J. Am. Acad. Dermatol. 2022, 87, 592–596. [Google Scholar] [CrossRef]
  74. Demehri, S.; Turkoz, A.; Manivasagam, S.; Yockey, L.J.; Turkoz, M.; Kopan, R. Elevated epidermal thymic stromal lymphopoietin levels establish an antitumor environment in the skin. Cancer Cell 2012, 22, 494–505. [Google Scholar] [CrossRef]
  75. Li, M.; Hener, P.; Zhang, Z.; Kato, S.; Metzger, D.; Chambon, P. Topical vitamin D3 and low-calcemic analogs induce thymic stromal lymphopoietin in mouse keratinocytes and trigger an atopic dermatitis. Proc. Natl. Acad. Sci. USA 2006, 103, 11736–11741. [Google Scholar] [CrossRef] [PubMed]
  76. Cunningham, T.J.; Tabacchi, M.; Eliane, J.P.; Tuchayi, S.M.; Manivasagam, S.; Mirzaalian, H.; Turkoz, A.; Kopan, R.; Schaffer, A.; Saavedra, A.P.; et al. Randomized trial of calcipotriol combined with 5-fluorouracil for skin cancer precursor immunotherapy. J. Clin. Investig. 2017, 127, 106–116. [Google Scholar] [CrossRef] [PubMed]
  77. Rosenberg, A.R.; Tabacchi, M.; Ngo, K.H.; Wallendorf, M.; Rosman, I.S.; Cornelius, L.A.; Demehri, S. Skin cancer precursor immunotherapy for squamous cell carcinoma prevention. JCI Insight 2019, 4, e125476. [Google Scholar] [CrossRef] [PubMed]
  78. Trodello, C.; Pepper, J.P.; Wong, M.; Wysong, A. Cisplatin and Cetuximab Treatment for Metastatic Cutaneous Squamous Cell Carcinoma: A Systematic Review. Dermatol. Surg. 2017, 43, 40–49. [Google Scholar] [CrossRef]
  79. Cowey, C.L.; Robert, N.J.; Espirito, J.L.; Davies, K.; Frytak, J.; Lowy, I.; Fury, M.G. Clinical outcomes among unresectable, locally advanced, and metastatic cutaneous squamous cell carcinoma patients treated with systemic therapy. Cancer Med. 2020, 9, 7381–7387. [Google Scholar] [CrossRef]
  80. Maubec, E.; Petrow, P.; Scheer-Senyarich, I.; Duvillard, P.; Lacroix, L.; Gelly, J.; Certain, A.; Duval, X.; Crickx, B.; Buffard, V.; et al. Phase II study of cetuximab as first-line single-drug therapy in patients with unresectable squamous cell carcinoma of the skin. J. Clin. Oncol. 2011, 29, 3419–3426. [Google Scholar] [CrossRef]
  81. Montaudie, H.; Viotti, J.; Combemale, P.; Dutriaux, C.; Dupin, N.; Robert, C.; Mortier, L.; Kaphan, R.; Duval-Modeste, A.B.; Dalle, S.; et al. Cetuximab is efficient and safe in patients with advanced cutaneous squamous cell carcinoma: A retrospective, multicentre study. Oncotarget 2020, 11, 378–385. [Google Scholar] [CrossRef] [PubMed]
  82. Di Nardo, L.; Pellegrini, C.; Di Stefani, A.; Del Regno, L.; Sollena, P.; Piccerillo, A.; Longo, C.; Garbe, C.; Fargnoli, M.C.; Peris, K. Molecular genetics of cutaneous squamous cell carcinoma: Perspective for treatment strategies. J. Eur. Acad. Dermatol. Venereol. 2020, 34, 932–941. [Google Scholar] [CrossRef]
  83. Picard, A.; Pedeutour, F.; Peyrade, F.; Saudes, L.; Duranton-Tanneur, V.; Chamorey, E.; Cardot-Leccia, N.; Sudaka, A.; Ettaiche, M.; Benchetrit, M.; et al. Association of Oncogenic Mutations in Patients with Advanced Cutaneous Squamous Cell Carcinomas Treated with Cetuximab. JAMA Dermatol. 2017, 153, 291–298. [Google Scholar] [CrossRef] [PubMed]
  84. Vaishampayan, P.; Curiel-Lewandrowski, C.; Dickinson, S.E. Review: PD-L1 as an emerging target in the treatment and prevention of keratinocytic skin cancer. Mol. Carcinog. 2023, 62, 52–61. [Google Scholar] [CrossRef] [PubMed]
  85. Ghidini, A.; Santangelo, D.; Vaccaro, G.; Chillura, M.; Petrelli, F. Cemiplimab in cutaneous squamous cell carcinomas (SCC): An overview and a clinical case. Oral Oncol. 2022, 128, 105847. [Google Scholar] [CrossRef] [PubMed]
  86. Burova, E.; Hermann, A.; Waite, J.; Potocky, T.; Lai, V.; Hong, S.; Liu, M.; Allbritton, O.; Woodruff, A.; Wu, Q.; et al. Characterization of the Anti-PD-1 Antibody REGN2810 and Its Antitumor Activity in Human PD-1 Knock-In Mice. Mol. Cancer Ther. 2017, 16, 861–870. [Google Scholar] [CrossRef] [PubMed]
  87. Rischin, D.; Migden, M.R.; Lim, A.M.; Schmults, C.D.; Khushalani, N.I.; Hughes, B.G.M.; Schadendorf, D.; Dunn, L.A.; Hernandez-Aya, L.; Chang, A.L.S.; et al. Phase 2 study of cemiplimab in patients with metastatic cutaneous squamous cell carcinoma: Primary analysis of fixed-dosing, long-term outcome of weight-based dosing. J. Immunother. Cancer 2020, 8, e000775. [Google Scholar] [CrossRef]
  88. Migden, M.R.; Khushalani, N.I.; Chang, A.L.S.; Lewis, K.D.; Schmults, C.D.; Hernandez-Aya, L.; Meier, F.; Schadendorf, D.; Guminski, A.; Hauschild, A.; et al. Cemiplimab in locally advanced cutaneous squamous cell carcinoma: Results from an open-label, phase 2, single-arm trial. Lancet Oncol. 2020, 21, 294–305. [Google Scholar] [CrossRef]
  89. Migden, M.R.; Rischin, D.; Schmults, C.D.; Guminski, A.; Hauschild, A.; Lewis, K.D.; Chung, C.H.; Hernandez-Aya, L.; Lim, A.M.; Chang, A.L.S.; et al. PD-1 Blockade with Cemiplimab in Advanced Cutaneous Squamous-Cell Carcinoma. N. Engl. J. Med. 2018, 379, 341–351. [Google Scholar] [CrossRef]
  90. Rios-Vinuela, E.; Alvarez, P.; Lavernia, J.; Serra-Guillen, C.; Requena, C.; Bernia, E.; Diago, A.; Llombart, B.; Sanmartin, O. Cemiplimab in Advanced Cutaneous Squamous Cell Carcinoma: Real-World Experience in a Monographic Oncology Center. Actas Dermosifiliogr. 2022, 113, T610–T615. [Google Scholar] [CrossRef]
  91. Baggi, A.; Quaglino, P.; Rubatto, M.; Depenni, R.; Guida, M.; Ascierto, P.A.; Trojaniello, C.; Queirolo, P.; Saponara, M.; Peris, K.; et al. Real world data of cemiplimab in locally advanced and metastatic cutaneous squamous cell carcinoma. Eur. J. Cancer 2021, 157, 250–258. [Google Scholar] [CrossRef] [PubMed]
  92. Strippoli, S.; Fanizzi, A.; Quaresmini, D.; Nardone, A.; Armenio, A.; Figliuolo, F.; Filotico, R.; Fucci, L.; Mele, F.; Traversa, M.; et al. Cemiplimab in an Elderly Frail Population of Patients with Locally Advanced or Metastatic Cutaneous Squamous Cell Carcinoma: A Single-Center Real-Life Experience from Italy. Front. Oncol. 2021, 11, 686308. [Google Scholar] [CrossRef]
  93. Hober, C.; Fredeau, L.; Pham-Ledard, A.; Boubaya, M.; Herms, F.; Celerier, P.; Aubin, F.; Beneton, N.; Dinulescu, M.; Jannic, A.; et al. Cemiplimab for Locally Advanced and Metastatic Cutaneous Squamous-Cell Carcinomas: Real-Life Experience from the French CAREPI Study Group. Cancers 2021, 13, 3547. [Google Scholar] [CrossRef] [PubMed]
  94. Kwok, G.; Yau, T.C.; Chiu, J.W.; Tse, E.; Kwong, Y.L. Pembrolizumab (Keytruda). Hum. Vaccines Immunother. 2016, 12, 2777–2789. [Google Scholar] [CrossRef]
  95. Grob, J.J.; Gonzalez, R.; Basset-Seguin, N.; Vornicova, O.; Schachter, J.; Joshi, A.; Meyer, N.; Grange, F.; Piulats, J.M.; Bauman, J.R.; et al. Pembrolizumab Monotherapy for Recurrent or Metastatic Cutaneous Squamous Cell Carcinoma: A Single-Arm Phase II Trial (KEYNOTE-629). J. Clin. Oncol. 2020, 38, 2916–2925. [Google Scholar] [CrossRef] [PubMed]
  96. Hughes, B.G.M.; Munoz-Couselo, E.; Mortier, L.; Bratland, A.; Gutzmer, R.; Roshdy, O.; Gonzalez Mendoza, R.; Schachter, J.; Arance, A.; Grange, F.; et al. Pembrolizumab for locally advanced and recurrent/metastatic cutaneous squamous cell carcinoma (KEYNOTE-629 study): An open-label, nonrandomized, multicenter, phase II trial. Ann. Oncol. 2021, 32, 1276–1285. [Google Scholar] [CrossRef] [PubMed]
  97. Maubec, E.; Boubaya, M.; Petrow, P.; Beylot-Barry, M.; Basset-Seguin, N.; Deschamps, L.; Grob, J.J.; Dreno, B.; Scheer-Senyarich, I.; Bloch-Queyrat, C.; et al. Phase II Study of Pembrolizumab As First-Line, Single-Drug Therapy for Patients with Unresectable Cutaneous Squamous Cell Carcinomas. J. Clin. Oncol. 2020, 38, 3051–3061. [Google Scholar] [CrossRef]
  98. William, W.N., Jr.; Feng, L.; Ferrarotto, R.; Ginsberg, L.; Kies, M.; Lippman, S.; Glisson, B.; Kim, E.S. Gefitinib for patients with incurable cutaneous squamous cell carcinoma: A single-arm phase II clinical trial. J. Am. Acad. Dermatol. 2017, 77, 1110–1113.e2. [Google Scholar] [CrossRef]
  99. Gold, K.A.; Kies, M.S.; William, W.N., Jr.; Johnson, F.M.; Lee, J.J.; Glisson, B.S. Erlotinib in the treatment of recurrent or metastatic cutaneous squamous cell carcinoma: A single-arm phase 2 clinical trial. Cancer 2018, 124, 2169–2173. [Google Scholar] [CrossRef]
  100. Jenni, D.; Karpova, M.B.; Muhleisen, B.; Mangana, J.; Dreier, J.; Hafner, J.; Dummer, R. A prospective clinical trial to assess lapatinib effects on cutaneous squamous cell carcinoma and actinic keratosis. ESMO Open 2016, 1, e000003. [Google Scholar] [CrossRef]
  101. Xu, G.; Fang, J.; Xu, J.; Shen, Z.; Huang, C.; Jiang, Y. Expression and significance of mammalian target of rapamycin in cutaneous squamous cell carcinoma and precancerous lesions. Bioengineered 2021, 12, 9930–9938. [Google Scholar] [CrossRef] [PubMed]
  102. Gluck, M.; Hodak, E.; Davidovici, B. Mammalian target of rapamycin inhibitors for prolonged secondary prevention of nonmelanoma skin cancer in solid organ transplant recipients. Dermatol. Ther. 2022, 35, e15649. [Google Scholar] [CrossRef] [PubMed]
  103. Salgo, R.; Gossmann, J.; Schofer, H.; Kachel, H.G.; Kuck, J.; Geiger, H.; Kaufmann, R.; Scheuermann, E.H. Switch to a sirolimus-based immunosuppression in long-term renal transplant recipients: Reduced rate of (pre-)malignancies and nonmelanoma skin cancer in a prospective, randomized, assessor-blinded, controlled clinical trial. Am. J. Transplant. 2010, 10, 1385–1393. [Google Scholar] [CrossRef] [PubMed]
  104. Song, Y.; Guerrero-Juarez, C.F.; Chen, Z.; Tang, Y.; Ma, X.; Lv, C.; Bi, X.; Deng, M.; Bu, L.; Tian, Y.; et al. The Msi1-mTOR pathway drives the pathogenesis of mammary and extramammary Paget’s disease. Cell Res. 2020, 30, 854–872. [Google Scholar] [CrossRef] [PubMed]
  105. Ansary, T.M.; Hossain, M.D.R.; Komine, M.; Ohtsuki, M. Immunotherapy for the Treatment of Squamous Cell Carcinoma: Potential Benefits and Challenges. Int. J. Mol. Sci. 2022, 23, 8530. [Google Scholar] [CrossRef] [PubMed]
  106. Munhoz, R.R.; Nader-Marta, G.; de Camargo, V.P.; Queiroz, M.M.; Cury-Martins, J.; Ricci, H.; de Mattos, M.R.; de Menezes, T.A.F.; Machado, G.U.C.; Bertolli, E.; et al. A phase 2 study of first-line nivolumab in patients with locally advanced or metastatic cutaneous squamous-cell carcinoma. Cancer 2022, 128, 4223–4231. [Google Scholar] [CrossRef] [PubMed]
  107. Clingan, P.; Ladwa, R.; Brungs, D.; Harris, D.L.; McGrath, M.; Arnold, S.; Coward, J.; Fourie, S.; Kurochkin, A.; Malan, D.R.; et al. Efficacy and safety of cosibelimab, an anti-PD-L1 antibody, in metastatic cutaneous squamous cell carcinoma. J. Immunother. Cancer 2023, 11, e007637. [Google Scholar] [CrossRef] [PubMed]
  108. Nguyen, T.A.; Offner, M.; Hamid, O.; Zumsteg, Z.S.; Gharavi, N.M. Complete and Sustained Remission of Metastatic Cutaneous Squamous Cell Carcinoma in a Liver Transplant Patient Treated with Talimogene Laherparepvec. Dermatol. Surg. 2021, 47, 820–822. [Google Scholar] [CrossRef] [PubMed]
  109. Zhang, T.; Jou, T.H.; Hsin, J.; Wang, Z.; Huang, K.; Ye, J.; Yin, H.; Xing, Y. Talimogene Laherparepvec (T-VEC): A Review of the Recent Advances in Cancer Therapy. J. Clin. Med. 2023, 12, 1098. [Google Scholar] [CrossRef]
  110. McDaniel, B.; Badri, T.; Steele, R.B. Basal Cell Carcinoma. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar]
  111. Cameron, M.C.; Lee, E.; Hibler, B.P.; Giordano, C.N.; Barker, C.A.; Mori, S.; Cordova, M.; Nehal, K.S.; Rossi, A.M. Basal cell carcinoma: Contemporary approaches to diagnosis, treatment, and prevention. J. Am. Acad. Dermatol. 2019, 80, 321–339. [Google Scholar] [CrossRef]
  112. Gallagher, R.P.; Ma, B.; McLean, D.I.; Yang, C.P.; Ho, V.; Carruthers, J.A.; Warshawski, L.M. Trends in basal cell carcinoma, squamous cell carcinoma, and melanoma of the skin from 1973 through 1987. J. Am. Acad. Dermatol. 1990, 23, 413–421. [Google Scholar] [CrossRef] [PubMed]
  113. Pranteda, G.; Grimaldi, M.; Lombardi, M.; Pranteda, G.; Arcese, A.; Cortesi, G.; Muscianese, M.; Bottoni, U. Basal cell carcinoma: Differences according to anatomic location and clinical-pathological subtypes. G. Ital. Dermatol. Venereol. 2014, 149, 423–426. [Google Scholar] [PubMed]
  114. Linos, E.; Chren, M.M. Active Surveillance as a Management Option for Low-risk Basal Cell Carcinoma. JAMA Intern. Med. 2021, 181, 1032–1033. [Google Scholar] [CrossRef] [PubMed]
  115. Kim, D.P.; Kus, K.J.B.; Ruiz, E. Basal Cell Carcinoma Review. Hematol. Oncol. Clin. N. Am. 2019, 33, 13–24. [Google Scholar] [CrossRef] [PubMed]
  116. Peris, K.; Licitra, L.; Ascierto, P.A.; Corvo, R.; Simonacci, M.; Picciotto, F.; Gualdi, G.; Pellacani, G.; Santoro, A. Identifying locally advanced basal cell carcinoma eligible for treatment with vismodegib: An expert panel consensus. Future Oncol. 2015, 11, 703–712. [Google Scholar] [CrossRef] [PubMed]
  117. Amici, J.M.; Battistella, M.; Beylot-Barry, M.; Chatellier, A.; Dalac-Ra, S.; Dreno, B.; Falandry, C.; Froget, N.; Giacchero, D.; Grob, J.J.; et al. Defining and recognising locally advanced basal cell carcinoma. Eur. J. Dermatol. 2015, 25, 586–594. [Google Scholar] [CrossRef] [PubMed]
  118. Gandhi, S.A.; Kampp, J. Skin Cancer Epidemiology, Detection, and Management. Med. Clin. N. Am. 2015, 99, 1323–1335. [Google Scholar] [CrossRef] [PubMed]
  119. Heath, M.S.; Bar, A. Basal Cell Carcinoma. Dermatol. Clin. 2023, 41, 13–21. [Google Scholar] [CrossRef] [PubMed]
  120. 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]
  121. Grob, J.J.; Gaudy-Marqueste, C.; Guminski, A.; Malvehy, J.; Basset-Seguin, N.; Bertrand, B.; Fernandez-Penas, P.; Kaufmann, R.; Zalaudek, I.; Fargnoli, M.C.; et al. Position statement on classification of basal cell carcinomas. Part 2: EADO proposal for new operational staging system adapted to basal cell carcinomas. J. Eur. Acad. Dermatol. Venereol. 2021, 35, 2149–2153. [Google Scholar] [CrossRef]
  122. Peris, K.; Fargnoli, M.C.; Kaufmann, R.; Arenberger, P.; Bastholt, L.; Seguin, N.B.; Bataille, V.; Brochez, L.; Del Marmol, V.; Dummer, R.; et al. European consensus-based interdisciplinary guideline for diagnosis and treatment of basal cell carcinoma-update 2023. Eur. J. Cancer 2023, 192, 113254. [Google Scholar] [CrossRef]
  123. Goldenberg, G.; Karagiannis, T.; Palmer, J.B.; Lotya, J.; O’Neill, C.; Kisa, R.; Herrera, V.; Siegel, D.M. Incidence and prevalence of basal cell carcinoma (BCC) and locally advanced BCC (LABCC) in a large commercially insured population in the United States: A retrospective cohort study. J. Am. Acad. Dermatol. 2016, 75, 957–966.e2. [Google Scholar] [CrossRef] [PubMed]
  124. Work, G.; Invited, R.; Kim, J.Y.S.; Kozlow, J.H.; Mittal, B.; Moyer, J.; Olencki, T.; Rodgers, P. Guidelines of care for the management of basal cell carcinoma. J. Am. Acad. Dermatol. 2018, 78, 540–559. [Google Scholar] [CrossRef]
  125. Trakatelli, M.; Morton, C.; Nagore, E.; Ulrich, C.; Del Marmol, V.; Peris, K.; Basset-Seguin, N.; Developed by the Guideline Subcommittee of the European Dermatology Forum. Update of the European guidelines for basal cell carcinoma management. Eur. J. Dermatol. 2014, 24, 312–329. [Google Scholar] [CrossRef] [PubMed]
  126. van Loo, E.; Mosterd, K.; Krekels, G.A.; Roozeboom, M.H.; Ostertag, J.U.; Dirksen, C.D.; Steijlen, P.M.; Neumann, H.A.; 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 10 year follow-up. Eur. J. Cancer 2014, 50, 3011–3020. [Google Scholar] [CrossRef] [PubMed]
  127. 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: A Systematic Review and Network Meta-analysis. Ann. Intern. Med. 2018, 169, 456–466. [Google Scholar] [CrossRef] [PubMed]
  128. Berking, C.; Hauschild, A.; Kolbl, O.; Mast, G.; Gutzmer, R. Basal cell carcinoma-treatments for the commonest skin cancer. Dtsch. Arztebl. Int. 2014, 111, 389–395. [Google Scholar] [CrossRef] [PubMed]
  129. Tinelli, M.; Ozolins, M.; Bath-Hextall, F.; Williams, H.C. What determines patient preferences for treating low risk basal cell carcinoma when comparing surgery vs imiquimod? A discrete choice experiment survey from the SINS trial. BMC Dermatol. 2012, 12, 19. [Google Scholar] [CrossRef] [PubMed]
  130. 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]
  131. Verzi, A.E.; Micali, G.; Lacarrubba, F. Line-Field Confocal Optical Coherence Tomography May Enhance Monitoring of Superficial Basal Cell Carcinoma Treated with Imiquimod 5% Cream: A Pilot Study. Cancers 2021, 13, 4913. [Google Scholar] [CrossRef]
  132. Bath-Hextall, F.; Ozolins, M.; Armstrong, S.J.; Colver, G.B.; Perkins, W.; Miller, P.S.; Williams, H.C.; Surgery versus Imiquimod for Nodular Superficial Basal Cell Carcinoma (SINS) Study Group. Surgical excision versus imiquimod 5% cream for nodular and superficial basal-cell carcinoma (SINS): A multicentre, non-inferiority, randomised controlled trial. Lancet Oncol. 2014, 15, 96–105. [Google Scholar] [CrossRef]
  133. Nanda, J.; Bermudez, R. Imiquimod. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar]
  134. Paoli, J.; Gyllencreutz, J.D.; Fougelberg, J.; Backman, E.J.; Modin, M.; Polesie, S.; Zaar, O. Nonsurgical Options for the Treatment of Basal Cell Carcinoma. Dermatol. Pract. Concept. 2019, 9, 75–81. [Google Scholar] [CrossRef] [PubMed]
  135. Arits, A.H.; Mosterd, K.; Essers, B.A.; 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] [PubMed]
  136. Bubna, A.K. Imiquimod—Its role in the treatment of cutaneous malignancies. Indian J. Pharmacol. 2015, 47, 354–359. [Google Scholar] [CrossRef]
  137. Tanese, K. Diagnosis and Management of Basal Cell Carcinoma. Curr. Treat. Opt. Oncol. 2019, 20, 13. [Google Scholar] [CrossRef] [PubMed]
  138. Jia, H.X.; He, Y.L. Efficacy and safety of imiquimod 5% cream for basal cell carcinoma: A meta-analysis of randomized controlled trial. J. Dermatol. Treat. 2020, 31, 831–838. [Google Scholar] [CrossRef]
  139. Shaw, F.M.; Weinstock, M.A. Comparing Topical Treatments for Basal Cell Carcinoma. J. Investig. Dermatol. 2018, 138, 484–486. [Google Scholar] [CrossRef]
  140. Jansen, M.H.E.; Mosterd, K.; Arits, A.; Roozeboom, M.H.; Sommer, A.; Essers, B.A.B.; van Pelt, H.P.A.; Quaedvlieg, P.J.F.; 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]
  141. Rimkus, T.K.; Carpenter, R.L.; Qasem, S.; Chan, M.; Lo, H.W. Targeting the Sonic Hedgehog Signaling Pathway: Review of Smoothened and GLI Inhibitors. Cancers 2016, 8, 22. [Google Scholar] [CrossRef]
  142. Sternfeld, A.; Rosenwasser-Weiss, S.; Ben-Yehuda, G.; Shefer, H.K.; Friedman-Gohas, M.; Yassur, I.; Tauber, G.; Bejar, J.; Olshinka, A.; Vardizer, Y.; et al. Gene-Related Response of Basal Cell Carcinoma to Biologic Treatment with Vismodegib. Sci. Rep. 2020, 10, 1244. [Google Scholar] [CrossRef]
  143. Ingham, P.W.; McMahon, A.P. Hedgehog signaling in animal development: Paradigms and principles. Genes Dev. 2001, 15, 3059–3087. [Google Scholar] [CrossRef] [PubMed]
  144. Tremblay, M.R.; Nevalainen, M.; Nair, S.J.; Porter, J.R.; Castro, A.C.; Behnke, M.L.; Yu, L.C.; Hagel, M.; White, K.; Faia, K.; et al. Semisynthetic cyclopamine analogues as potent and orally bioavailable hedgehog pathway antagonists. J. Med. Chem. 2008, 51, 6646–6649. [Google Scholar] [CrossRef] [PubMed]
  145. Chen, J.K.; Taipale, J.; Cooper, M.K.; Beachy, P.A. Inhibition of Hedgehog signaling by direct binding of cyclopamine to Smoothened. Genes Dev. 2002, 16, 2743–2748. [Google Scholar] [CrossRef] [PubMed]
  146. Xie, P.; Lefrancois, P. Efficacy, safety, and comparison of sonic hedgehog inhibitors in basal cell carcinomas: A systematic review and meta-analysis. J. Am. Acad. Dermatol. 2018, 79, 1089–1100.e17. [Google Scholar] [CrossRef] [PubMed]
  147. Abe, Y.; Tanaka, N. Roles of the Hedgehog Signaling Pathway in Epidermal and Hair Follicle Development, Homeostasis, and Cancer. J. Dev. Biol. 2017, 5, 12. [Google Scholar] [CrossRef] [PubMed]
  148. 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]
  149. Atwood, S.X.; Sarin, K.Y.; Whitson, R.J.; Li, J.R.; Kim, G.; Rezaee, M.; Ally, M.S.; Kim, J.; Yao, C.; Chang, A.L.; et al. Smoothened variants explain the majority of drug resistance in basal cell carcinoma. Cancer Cell 2015, 27, 342–353. [Google Scholar] [CrossRef]
  150. 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]
  151. Scalvenzi, M.; Costa, C.; Cappello, M.; Villani, A. Reply to Woltsche N. et al. Managing adverse effects by dose reduction during routine treatment of locally advanced basal cell carcinoma with the hedgehog inhibitor vismodegib: A single-centre experience. J. Eur. Acad. Dermatol. Venereol. 2019, 33, e145–e147. [Google Scholar] [CrossRef]
  152. Basset-Seguin, N.; Hauschild, A.; Kunstfeld, R.; Grob, J.; Dreno, B.; Mortier, L.; Ascierto, P.A.; Licitra, L.; Dutriaux, C.; Thomas, L.; et al. Vismodegib in patients with advanced basal cell carcinoma: Primary analysis of STEVIE, an international, open-label trial. Eur. J. Cancer 2017, 86, 334–348. [Google Scholar] [CrossRef]
  153. Sekulic, A.; Migden, M.R.; Basset-Seguin, N.; Garbe, C.; Gesierich, A.; Lao, C.D.; Miller, C.; Mortier, L.; Murrell, D.F.; Hamid, O.; et al. Long-term safety and efficacy of vismodegib in patients with advanced basal cell carcinoma: Final update of the pivotal ERIVANCE BCC study. BMC Cancer 2017, 17, 332. [Google Scholar] [CrossRef] [PubMed]
  154. Danial, C.; Sarin, K.Y.; Oro, A.E.; Chang, A.L. An Investigator-Initiated Open-Label Trial of Sonidegib in Advanced Basal Cell Carcinoma Patients Resistant to Vismodegib. Clin. Cancer Res. 2016, 22, 1325–1329. [Google Scholar] [CrossRef] [PubMed]
  155. Villani, A.; Fabbrocini, G.; Costa, C.; Scalvenzi, M. Sonidegib: Safety and Efficacy in Treatment of Advanced Basal Cell Carcinoma. Dermatol. Ther. 2020, 10, 401–412. [Google Scholar] [CrossRef] [PubMed]
  156. Villani, A.; Fabbrocini, G.; Costa, C.; Ocampo-Garza, S.S.; Lallas, A.; Scalvenzi, M. Expert opinion on sonidegib efficacy, safety and tolerability. Expert Opin. Drug Saf. 2021, 20, 877–882. [Google Scholar] [CrossRef] [PubMed]
  157. 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] [PubMed]
  158. Dummer, R.; Guminksi, A.; Gutzmer, R.; Lear, J.T.; Lewis, K.D.; Chang, A.L.S.; 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. 2020, 182, 1369–1378. [Google Scholar] [CrossRef] [PubMed]
  159. Villani, A.; Ocampo-Garza, S.S.; Potestio, L.; Fabbrocini, G.; Ocampo-Candiani, J.; Ocampo-Garza, J.; Scalvenzi, M. Cemiplimab for the treatment of advanced cutaneous squamous cell carcinoma. Expert Opin. Drug Saf. 2022, 21, 21–29. [Google Scholar] [CrossRef] [PubMed]
  160. Chang, J.; Zhu, G.A.; Cheung, C.; Li, S.; Kim, J.; Chang, A.L. Association between Programmed Death Ligand 1 Expression in Patients with Basal Cell Carcinomas and the Number of Treatment Modalities. JAMA Dermatol. 2017, 153, 285–290. [Google Scholar] [CrossRef] [PubMed]
  161. Jayaraman, S.S.; Rayhan, D.J.; Hazany, S.; Kolodney, M.S. Mutational landscape of basal cell carcinomas by whole-exome sequencing. J. Investig. Dermatol. 2014, 134, 213–220. [Google Scholar] [CrossRef]
  162. 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]
  163. Jimeno, A.; Weiss, G.J.; Miller, W.H., Jr.; Gettinger, S.; Eigl, B.J.; Chang, A.L.; Dunbar, J.; Devens, S.; Faia, K.; Skliris, G.; et al. Phase I study of the Hedgehog pathway inhibitor IPI-926 in adult patients with solid tumors. Clin. Cancer Res. 2013, 19, 2766–2774. [Google Scholar] [CrossRef]
  164. 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-Naive and Previously Treated Basal Cell Carcinoma. Clin. Cancer Res. 2018, 24, 2082–2091. [Google Scholar] [CrossRef]
  165. Ueno, H.; Kondo, S.; Yoshikawa, S.; Inoue, K.; Andre, V.; Tajimi, M.; Murakami, H. A phase I and pharmacokinetic study of taladegib, a Smoothened inhibitor, in Japanese patients with advanced solid tumors. Investig. New Drugs 2018, 36, 647–656. [Google Scholar] [CrossRef] [PubMed]
  166. 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] [PubMed]
  167. Nirenberg, A.; Steinman, H.; Dixon, J.; Dixon, A. Merkel cell carcinoma update: The case for two tumours. J. Eur. Acad. Dermatol. Venereol. 2020, 34, 1425–1431. [Google Scholar] [CrossRef]
  168. Mistry, K.; Levell, N.J.; Craig, P.; Steven, N.M.; Venables, Z.C. Merkel cell carcinoma. Skin Health Dis. 2021, 1, e55. [Google Scholar] [CrossRef]
  169. Heath, M.; Jaimes, N.; Lemos, B.; Mostaghimi, A.; Wang, L.C.; Penas, P.F.; Nghiem, P. Clinical characteristics of Merkel cell carcinoma at diagnosis in 195 patients: The AEIOU features. J. Am. Acad. Dermatol. 2008, 58, 375–381. [Google Scholar] [CrossRef]
  170. Harms, K.L.; Healy, M.A.; Nghiem, P.; Sober, A.J.; Johnson, T.M.; Bichakjian, C.K.; Wong, S.L. Analysis of Prognostic Factors from 9387 Merkel Cell Carcinoma Cases Forms the Basis for the New 8th Edition AJCC Staging System. Ann. Surg. Oncol. 2016, 23, 3564–3571. [Google Scholar] [CrossRef]
  171. Bichakjian, C.K.; Olencki, T.; Aasi, S.Z.; Alam, M.; Andersen, J.S.; Blitzblau, R.; Bowen, G.M.; Contreras, C.M.; Daniels, G.A.; Decker, R.; et al. Merkel Cell Carcinoma, Version 1.2018, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2018, 16, 742–774. [Google Scholar] [CrossRef] [PubMed]
  172. Gauci, M.L.; Aristei, C.; Becker, J.C.; Blom, A.; Bataille, V.; Dreno, B.; Del Marmol, V.; Forsea, A.M.; Fargnoli, M.C.; Grob, J.J.; et al. Diagnosis and treatment of Merkel cell carcinoma: European consensus-based interdisciplinary guideline—Update 2022. Eur. J. Cancer 2022, 171, 203–231. [Google Scholar] [CrossRef]
  173. Perez, M.C.; de Pinho, F.R.; Holstein, A.; Oliver, D.E.; Naqvi, S.M.H.; Kim, Y.; Messina, J.L.; Burke, E.; Gonzalez, R.J.; Sarnaik, A.A.; et al. Resection Margins in Merkel Cell Carcinoma: Is a 1-cm Margin Wide Enough? Ann. Surg. Oncol. 2018, 25, 3334–3340. [Google Scholar] [CrossRef] [PubMed]
  174. Park, S.Y.; Doolittle-Amieva, C.; Moshiri, Y.; Akaike, T.; Parvathaneni, U.; Bhatia, S.; Zaba, L.C.; Nghiem, P. How we treat Merkel cell carcinoma: Within and beyond current guidelines. Future Oncol. 2021, 17, 1363–1377. [Google Scholar] [CrossRef] [PubMed]
  175. Schmults, C.D.; Blitzblau, R.; Aasi, S.Z.; Alam, M.; Amini, A.; Bibee, K.; Bolotin, D.; Bordeaux, J.; Chen, P.L.; Contreras, C.M.; et al. NCCN Guidelines(R) Insights: Merkel Cell Carcinoma, Version 1.2024. J. Natl. Compr. Cancer Netw. 2024, 22, e240002. [Google Scholar] [CrossRef] [PubMed]
  176. Harrington, C.; Kwan, W. Outcomes of Merkel cell carcinoma treated with radiotherapy without radical surgical excision. Ann. Surg. Oncol. 2014, 21, 3401–3405. [Google Scholar] [CrossRef] [PubMed]
  177. D’Angelo, S.P.; Lebbe, C.; Mortier, L.; Brohl, A.S.; Fazio, N.; Grob, J.J.; Prinzi, N.; Hanna, G.J.; Hassel, J.C.; Kiecker, F.; et al. First-line avelumab in a cohort of 116 patients with metastatic Merkel cell carcinoma (JAVELIN Merkel 200): Primary and biomarker analyses of a phase II study. J. Immunother. Cancer 2021, 9, e002646. [Google Scholar] [CrossRef] [PubMed]
  178. Levy, S.; Aarts, M.J.B.; Eskens, F.; Keymeulen, K.; Been, L.B.; Grunhagen, D.; van Akkooi, A.; Jalving, M.; Tesselaar, M.E.T. Avelumab for advanced Merkel cell carcinoma in The Netherlands: A real-world cohort. J. Immunother. Cancer 2020, 8, e001076. [Google Scholar] [CrossRef]
  179. Cowey, C.L.; Liu, F.X.; Kim, R.; Boyd, M.; Fulcher, N.; Krulewicz, S.; Kasturi, V.; Bhanegaonkar, A. Real-world clinical outcomes with first-line avelumab in locally advanced/metastatic Merkel cell carcinoma in the USA: SPEAR-Merkel. Future Oncol. 2021, 17, 2339–2350. [Google Scholar] [CrossRef] [PubMed]
  180. Nghiem, P.T.; Bhatia, S.; Lipson, E.J.; Kudchadkar, R.R.; Miller, N.J.; Annamalai, L.; Berry, S.; Chartash, E.K.; Daud, A.; Fling, S.P.; et al. PD-1 Blockade with Pembrolizumab in Advanced Merkel-Cell Carcinoma. N. Engl. J. Med. 2016, 374, 2542–2552. [Google Scholar] [CrossRef] [PubMed]
  181. Nghiem, P.; Bhatia, S.; Lipson, E.J.; Sharfman, W.H.; Kudchadkar, R.R.; Brohl, A.S.; Friedlander, P.A.; Daud, A.; Kluger, H.M.; Reddy, S.A.; et al. Durable Tumor Regression and Overall Survival in Patients with Advanced Merkel Cell Carcinoma Receiving Pembrolizumab as First-Line Therapy. J. Clin Oncol 2019, 37, 693–702. [Google Scholar] [CrossRef]
  182. Tarabadkar, E.S.; Thomas, H.; Blom, A.; Parvathaneni, U.; Olencki, T.; Nghiem, P.; Bhatia, S. Clinical Benefit from Tyrosine Kinase Inhibitors in Metastatic Merkel Cell Carcinoma: A Case Series of 5 Patients. Am. J. Case Rep. 2018, 19, 505–511. [Google Scholar] [CrossRef]
  183. Dresang, L.R.; Guastafierro, A.; Arora, R.; Normolle, D.; Chang, Y.; Moore, P.S. Response of Merkel cell polyomavirus-positive merkel cell carcinoma xenografts to a survivin inhibitor. PLoS ONE 2013, 8, e80543. [Google Scholar] [CrossRef]
  184. Verhaegen, M.E.; Mangelberger, D.; Weick, J.W.; Vozheiko, T.D.; Harms, P.W.; Nash, K.T.; Quintana, E.; Baciu, P.; Johnson, T.M.; Bichakjian, C.K.; et al. Merkel cell carcinoma dependence on bcl-2 family members for survival. J. Investig. Dermatol. 2014, 134, 2241–2250. [Google Scholar] [CrossRef] [PubMed]
  185. Ananthapadmanabhan, V.; Frost, T.C.; Soroko, K.M.; Knott, A.; Magliozzi, B.J.; Gokhale, P.C.; Tirunagaru, V.G.; Doebele, R.C.; DeCaprio, J.A. Milademetan is a highly potent MDM2 inhibitor in Merkel cell carcinoma. JCI Insight 2022, 7, e160513. [Google Scholar] [CrossRef]
  186. Houben, R.; Dreher, C.; Angermeyer, S.; Borst, A.; Utikal, J.; Haferkamp, S.; Peitsch, W.K.; Schrama, D.; Hesbacher, S. Mechanisms of p53 restriction in Merkel cell carcinoma cells are independent of the Merkel cell polyoma virus T antigens. J. Investig. Dermatol. 2013, 133, 2453–2460. [Google Scholar] [CrossRef] [PubMed]
  187. Gardair, C.; Samimi, M.; Touze, A.; Coursaget, P.; Lorette, G.; Caille, A.; Wierzbicka, E.; Croue, A.; Avenel-Audran, M.; Aubin, F.; et al. Somatostatin Receptors 2A and 5 Are Expressed in Merkel Cell Carcinoma with No Association with Disease Severity. Neuroendocrinology 2015, 101, 223–235. [Google Scholar] [CrossRef] [PubMed]
  188. Akaike, T.; Qazi, J.; Anderson, A.; Behnia, F.S.; Shinohara, M.M.; Akaike, G.; Hippe, D.S.; Thomas, H.; Takagishi, S.R.; Lachance, K.; et al. High somatostatin receptor expression and efficacy of somatostatin analogues in patients with metastatic Merkel cell carcinoma. Br. J. Dermatol. 2021, 184, 319–327. [Google Scholar] [CrossRef] [PubMed]
  189. Martini, M.; De Santis, M.C.; Braccini, L.; Gulluni, F.; Hirsch, E. PI3K/AKT signaling pathway and cancer: An updated review. Ann. Med. 2014, 46, 372–383. [Google Scholar] [CrossRef] [PubMed]
  190. Hafner, C.; Houben, R.; Baeurle, A.; Ritter, C.; Schrama, D.; Landthaler, M.; Becker, J.C. Activation of the PI3K/AKT pathway in Merkel cell carcinoma. PLoS ONE 2012, 7, e31255. [Google Scholar] [CrossRef]
  191. Nardi, V.; Song, Y.; Santamaria-Barria, J.A.; Cosper, A.K.; Lam, Q.; Faber, A.C.; Boland, G.M.; Yeap, B.Y.; Bergethon, K.; Scialabba, V.L.; et al. Activation of PI3K signaling in Merkel cell carcinoma. Clin. Cancer Res. 2012, 18, 1227–1236. [Google Scholar] [CrossRef]
  192. Lin, Z.; McDermott, A.; Shao, L.; Kannan, A.; Morgan, M.; Stack, B.C., Jr.; Moreno, M.; Davis, D.A.; Cornelius, L.A.; Gao, L. Chronic mTOR activation promotes cell survival in Merkel cell carcinoma. Cancer Lett. 2014, 344, 272–281. [Google Scholar] [CrossRef]
  193. Kannan, A.; Lin, Z.; Shao, Q.; Zhao, S.; Fang, B.; Moreno, M.A.; Vural, E.; Stack, B.C., Jr.; Suen, J.Y.; Kannan, K.; et al. Dual mTOR inhibitor MLN0128 suppresses Merkel cell carcinoma (MCC) xenograft tumor growth. Oncotarget 2016, 7, 6576–6592. [Google Scholar] [CrossRef] [PubMed]
  194. Sundqvist, B.; Kilpinen, S.; Bohling, T.; Koljonen, V.; Sihto, H. Activation of Oncogenic and Immune-Response Pathways Is Linked to Disease-Specific Survival in Merkel Cell Carcinoma. Cancers 2022, 14, 3591. [Google Scholar] [CrossRef] [PubMed]
  195. Shiver, M.B.; Mahmoud, F.; Gao, L. Response to Idelalisib in a Patient with Stage IV Merkel-Cell Carcinoma. N. Engl. J. Med. 2015, 373, 1580–1582. [Google Scholar] [CrossRef] [PubMed]
  196. Fang, B.; Kannan, A.; Zhao, S.; Nguyen, Q.H.; Ejadi, S.; Yamamoto, M.; Camilo Barreto, J.; Zhao, H.; Gao, L. Inhibition of PI3K by copanlisib exerts potent antitumor effects on Merkel cell carcinoma cell lines and mouse xenografts. Sci. Rep. 2020, 10, 8867. [Google Scholar] [CrossRef] [PubMed]
  197. Chteinberg, E.; Wetzels, S.; Gerritsen, W.; Temmerman, L.; van den Oord, J.; Biessen, E.; Kurz, A.K.; Winnepenninckx, V.; Zenke, M.; Speel, E.J.; et al. Navitoclax combined with Alpelisib effectively inhibits Merkel cell carcinoma cell growth in vitro. Ther. Adv. Med. Oncol. 2020, 12, 1758835920975621. [Google Scholar] [CrossRef]
  198. Burrows, J.N.; Chibale, K.; Wells, T.N. The state of the art in anti-malarial drug discovery and development. Curr. Top. Med. Chem. 2011, 11, 1226–1254. [Google Scholar] [CrossRef]
  199. Sarma, B.; Willmes, C.; Angerer, L.; Adam, C.; Becker, J.C.; Kervarrec, T.; Schrama, D.; Houben, R. Artesunate Affects T Antigen Expression and Survival of Virus-Positive Merkel Cell Carcinoma. Cancers 2020, 12, 919. [Google Scholar] [CrossRef] [PubMed]
  200. Kufe, D.W. MUC1-C in chronic inflammation and carcinogenesis; emergence as a target for cancer treatment. Carcinogenesis 2020, 41, 1173–1183. [Google Scholar] [CrossRef] [PubMed]
  201. Morimoto, Y.; Fushimi, A.; Yamashita, N.; Hagiwara, M.; Bhattacharya, A.; Cheng, J.; Frost, T.C.; Ahmad, R.; Daimon, T.; Huang, L.; et al. Addiction of Merkel cell carcinoma to MUC1-C identifies a potential new target for treatment. Oncogene 2022, 41, 3511–3523. [Google Scholar] [CrossRef]
  202. Glutsch, V.; Kneitz, H.; Gesierich, A.; Goebeler, M.; Haferkamp, S.; Becker, J.C.; Ugurel, S.; Schilling, B. Activity of ipilimumab plus nivolumab in avelumab-refractory Merkel cell carcinoma. Cancer Immunol. Immunother. 2021, 70, 2087–2093. [Google Scholar] [CrossRef]
  203. Khaddour, K.; Rosman, I.S.; Dehdashti, F.; Ansstas, G. Durable remission after rechallenge with ipilimumab and nivolumab in metastatic Merkel cell carcinoma refractory to avelumab: Any role for sequential immunotherapy? J. Dermatol. 2021, 48, e80–e81. [Google Scholar] [CrossRef] [PubMed]
  204. Shiravand, Y.; Khodadadi, F.; Kashani, S.M.A.; Hosseini-Fard, S.R.; Hosseini, S.; Sadeghirad, H.; Ladwa, R.; O’Byrne, K.; Kulasinghe, A. Immune Checkpoint Inhibitors in Cancer Therapy. Curr. Oncol. 2022, 29, 3044–3060. [Google Scholar] [CrossRef] [PubMed]
  205. Joshi, T.P.; Farr, M.A.; Hsiou, D.A.; Nugent, S.; Fathy, R.A.; Lewis, D.J. Therapeutic targets for vaccination in polyomavirus-driven Merkel cell carcinoma. Dermatol. Ther. 2022, 35, e15580. [Google Scholar] [CrossRef] [PubMed]
  206. Westbrook, B.C.; Norwood, T.G.; Terry, N.L.J.; McKee, S.B.; Conry, R.M. Talimogene laherparepvec induces durable response of regionally advanced Merkel cell carcinoma in 4 consecutive patients. JAAD Case Rep. 2019, 5, 782–786. [Google Scholar] [CrossRef] [PubMed]
  207. Cilento, M.A.; Klein, O.; Egan, E.; Roberts-Thomson, R. Talimogene laherparepvec resulting in near-complete response in a patient with treatment-refractory Merkel cell carcinoma. Australas. J. Dermatol. 2022, 63, e222–e225. [Google Scholar] [CrossRef] [PubMed]
  208. Casale, F.; Tchanque-Fossuo, C.; Stepenaskie, S.; Durkin, J. Complete response of Merkel cell carcinoma with talimogene laherparepvec (TVEC) monotherapy. Dermatol. Online J. 2022, 28, 5. [Google Scholar] [CrossRef] [PubMed]
  209. Nguyen, M.H.K.; Leong, S.P.; Abendroth, R.; Kashani-Sabet, M.; Kim, K.B. Complete clinical response to intralesional talimogene laherparepvec injection in a patient with recurrent, regionally advanced Merkel cell carcinoma. JAAD Case Rep. 2019, 5, 849–851. [Google Scholar] [CrossRef]
  210. Nahm, W.J.; Shen, J.; Zito, P.M.; Gonzalez, A.M.; Nagrani, N.; Moore, K.; Badiavas, E.V.; Kirsner, R.S.; Nichols, A.J. A Non-Surgical and Cost-Effective Treatment Approach Employing Topical Imiquimod, 5-Fluorouracil, and Tretinoin for Primary Non-Melanoma Skin Cancers. J. Drugs Dermatol. 2021, 20, 260–267. [Google Scholar] [CrossRef]
Figure 1. Simplified algorithm for the treatment of cSCC. cSCC, cutaneous squamous cell carcinoma; MCS, micrographically controlled surgery; RT, radiotherapy; 5-FU, 5-fluorouracil; PDT, photodynamic therapy.
Figure 1. Simplified algorithm for the treatment of cSCC. cSCC, cutaneous squamous cell carcinoma; MCS, micrographically controlled surgery; RT, radiotherapy; 5-FU, 5-fluorouracil; PDT, photodynamic therapy.
Ijms 25 07056 g001
Figure 2. Simplified algorithm for the treatment of BCC. BCC, basal cell carcinoma; MCS, micrographically controlled surgery; RT, radiotherapy; 5-FU, 5-fluorouracil; PDT, photodynamic therapy.
Figure 2. Simplified algorithm for the treatment of BCC. BCC, basal cell carcinoma; MCS, micrographically controlled surgery; RT, radiotherapy; 5-FU, 5-fluorouracil; PDT, photodynamic therapy.
Ijms 25 07056 g002
Figure 3. Simplified algorithm for the treatment of MCC. MCC, Merkel cell carcinoma; LN, lymph node; SLNB, sentinel lymph node biopsy; CLND complete lymph node dissection; RT, radiotherapy; PET, positron emission tomography; CT, computed tomography.
Figure 3. Simplified algorithm for the treatment of MCC. MCC, Merkel cell carcinoma; LN, lymph node; SLNB, sentinel lymph node biopsy; CLND complete lymph node dissection; RT, radiotherapy; PET, positron emission tomography; CT, computed tomography.
Ijms 25 07056 g003
Table 1. Ongoing trials in cutaneous squamous cell carcinoma (cSCC).
Table 1. Ongoing trials in cutaneous squamous cell carcinoma (cSCC).
NCT IdentifierStudy TitleConditionsPhasesPatients
Topical TreatmentNCT00652080An Open-Label Safety Study of API 31510 in a Topical Cream for in Situ Cutaneous Squamous Cell Carcinoma (SCCIS)In situ cSCCPHASE1/235
EGFR InhibitorNCT02324608Cetuximab Before Surgery in Treating Patients With Aggressive Locally Advanced Skin CancerLocally advanced cSCCNA15
NCT00126555Gefitinib in Treating Patients Who Are Undergoing Surgery and/or Radiation Therapy for Locally Advanced or Recurrent Squamous Cell Skin CancerLocally advanced cSCC or Recurrent cSCCPHASE223
NCT01198028Erlotinib in Treating Patients With Recurrent or Metastatic Skin Squamous Cell CarcinomaRecurrent or Metastatic cSCCPHASE242
ImmunotherapyNCT02883556Study of Pembrolizumab as First Line Therapy in Patients With Unresectable Squamous Cell Carcinoma of the SkinUnresectable cSCCPHASE257
NCT03834233Nivolumab in Patients With Advanced Cutaneous Squamous Cell CarcinomaAdvanced cSCCPHASE224
NCT03212404Phase 1 Study of CK-301 (Cosibelimab) as a Single Agent in Subjects With Advanced CancersAdvanced cSCCPHASE1500
NCT03714828Study of TVEC in Patients With Cutaneous Squamous Cell CancercSCCPHASE211
NCT04163952Talimogene Laherparepvec and Panitumumab for the Treatment of Locally Advanced or Metastatic Squamous Cell Carcinoma of the SkinLocally advanced cSCC or Recurrent cSCCPHASE15
NCT02760498Study of REGN2810 in Patients With Advanced Cutaneous Squamous Cell CarcinomaAdvanced cSCCPHASE2432
Table 2. Ongoing trials in basal cell carcinoma (BCC).
Table 2. Ongoing trials in basal cell carcinoma (BCC).
NCT IdentifierStudy TitleConditionsPhasesPatients
Topical TreatmentNCT05157763A Study to Evaluate the Safety and Efficacy of EscharEx (EX-02) in the Treatment of Basal Cell CarcinomaSuperficial or nodular BCPHASE1/232
NCT00604890Dose-Ranging Clinical Trial of Topical Creams Containing API 31510 for the Treatment of Superficial Basal Cell CarcinomaSuperficial BCCPHASE1/2186
NCT03180528Topical Remetinostat in Treating Patient With Cutaneous Basal Cell CancerBCCPHASE230
HH InhibitorsNCT02667574Study Evaluating the Interest of Vismodegib as Neo-adjuvant Treatment of Basal Cell Carcinoma (BCC)BCCPHASE255
NCT04806646Tailored Sonidegib Schedule After Complete Response in BCClaBCCPHASE221
NCT02690948Pembrolizumab With or Without Vismodegib in Treating Metastatic or Unresectable Basal Cell Skin CancerMetastatic or unresectable BCCPHASE1/216
NCT06344052To Assess the Safety and Efficacy of SP-002 With Vismodegib for the Treatment of Locally Advanced Basal Cell CarcinomaLocally advanced BCCPHASE280
NCT03035188Neoadjuvant Vismodegib in Patients With Large and/or Recurrent Resectable Basal Cell CarcinomaBCCPHASE240
NCT03972748Use Of Oral Itraconazole In Patients With Locally Limited Basocellular Carcinoma Of Skin.BCCNA28
NCT02735356Topical Itraconazole in Treating Patients With Basal Cell CancerBCCEARLY
PHASE1
9
NCT02828111Clinical Trial of Patidegib Gel 2%, 4%, and Vehicle Applied Once or Twice Daily to Decrease the GLI1 Biomarker in Sporadic Nodular Basal Cell CarcinomasNodular BCCPHASE236
NCT01700049Study Evaluating the Efficacy of Oral Vismodegib in Various Histologic SubtypesBCCPHASE228
ImmunotherapyNCT01327053A Phase II Study of Efficacy and Safety in Patients With Locally Advanced or Metastatic Basal Cell CarcinomaLocally advanced or metastatic BCCPHASE2230
NCT04679480Anti-PD1-antibody and Pulsed HHI for Advanced BCCAdvanced BCCPHASE220
NCT03521830Nivolumab Alone or Plus Relatlimab or Ipilimumab for Patients With Locally-Advanced Unresectable or Metastatic Basal Cell CarcinomaMetastatic or unresectable BCCPHASE257
NCT03132636PD-1 in Patients With Advanced Basal Cell Carcinoma Who Experienced Progression of Disease on Hedgehog Pathway Inhibitor Therapy, or Were Intolerant of Prior Hedgehog Pathway Inhibitor TherapyBCCPHASE2138
Table 3. Ongoing trials in Merkel cell carcinoma (MCC).
Table 3. Ongoing trials in Merkel cell carcinoma (MCC).
NCT IdentifierStudy TitleConditionsPhasesPatients
Targeted TherapiesNCT04869137Neoadjuvant Lenvatinib Plus Pembrolizumab in Merkel Cell CarcinomaMCCPHASE226
NCT03787602Navtemadlin (KRT-232) with or without Anti-PD-1/Anti-PD-L1 for the Treatment of Patients with Merkel Cell CarcinomaMCCPHASE1/2115
NCT02514824MLN0128 in Recurrent/Metastatic Merkel Cell CarcinomaRecurrent or
metastatic MCC
PHASE1|29
ImmunotherapyNCT04291885Immunotherapy Adjuvant Trial in Patients with Stage I-III Merkel Cell CarcinomaMCCPHASE2132
NCT05496036Neoadjuvant PD-1 Blockade in Resectable Merkel Cell CarcinomaMCCPHASE215
NCT06151236Neoadjuvant Nivolumab and Relatlimab in Merkel Cell CarcinomaMCCPHASE220
NCT03599713A Study of INCMGA00012 in Metastatic Merkel Cell Carcinoma (POD1UM-201)Metastatic MCCPHASE2107
NCT02488759An Investigational Immuno-therapy Study to Investigate the Safety and Effectiveness of Nivolumab, and Nivolumab Combination Therapy in Virus-associated TumorsAdvanced MCCPHASE1/2578
NCT03458117T-VEC in Non-melanoma Skin CancerMCCPHASE126
NCT02819843A Study of T-VEC (Talimogene Laherparepvec) with or without Radiotherapy for Melanoma, Merkel Cell Carcinoma, or Other Solid TumorsMCCPHASE219
NCT02978625Talimogene Laherparepvec and Nivolumab in Treating Patients with Refractory Lymphomas or Advanced or Refractory Non-melanoma Skin CancersMCCPHASE268
NCT02155647Avelumab in Participants with Merkel Cell Carcinoma (JAVELIN Merkel 200)MCCPHASE2204
NCT04393753Domatinostat in Combination with Avelumab in Patients with Advanced Merkel Cell Carcinoma Progressing on Anti-PD-(L)1Advanced MCCPHASE219
NCT05422781Study to Evaluate The Safety, Tolerability and Immunogenicity of 4 mg of ITI-3000 In Patients with Polyomavirus-Positive Merkel Cell Carcinoma (MCC)VP-MCCPHASE16
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sol, S.; Boncimino, F.; Todorova, K.; Waszyn, S.E.; Mandinova, A. Therapeutic Approaches for Non-Melanoma Skin Cancer: Standard of Care and Emerging Modalities. Int. J. Mol. Sci. 2024, 25, 7056. https://doi.org/10.3390/ijms25137056

AMA Style

Sol S, Boncimino F, Todorova K, Waszyn SE, Mandinova A. Therapeutic Approaches for Non-Melanoma Skin Cancer: Standard of Care and Emerging Modalities. International Journal of Molecular Sciences. 2024; 25(13):7056. https://doi.org/10.3390/ijms25137056

Chicago/Turabian Style

Sol, Stefano, Fabiana Boncimino, Kristina Todorova, Sarah Elizabeth Waszyn, and Anna Mandinova. 2024. "Therapeutic Approaches for Non-Melanoma Skin Cancer: Standard of Care and Emerging Modalities" International Journal of Molecular Sciences 25, no. 13: 7056. https://doi.org/10.3390/ijms25137056

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