Next Article in Journal
Regulation of Interleukin-36γ/IL-36R Signaling Axis by PIN1 in Epithelial Cell Transformation and Breast Tumorigenesis
Next Article in Special Issue
Advances in Management and Therapeutics of Cutaneous Basal Cell Carcinoma
Previous Article in Journal
The Prediction of Cardiac Events Using Contemporary Risk Prediction Models after Radiation Therapy for Head and Neck Cancer
Previous Article in Special Issue
Risk Factors and Clinicopathological Features for Developing a Subsequent Primary Cutaneous Squamous and Basal Cell Carcinomas
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advances in Cutaneous Squamous Cell Carcinoma Management

1
Department of Dermatology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
2
Department of Dermatology, University of Texas McGovern Medical School at Houston, 6655 Travis St. Suite 700, Houston, TX 77030, USA
3
Departments of Dermatology and Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(15), 3653; https://doi.org/10.3390/cancers14153653
Submission received: 30 June 2022 / Revised: 16 July 2022 / Accepted: 25 July 2022 / Published: 27 July 2022
(This article belongs to the Special Issue Research Progress of Cutaneous Squamous and Basal Cell Carcinomas)

Abstract

:

Simple Summary

Cutaneous squamous cell carcinoma (cSCC) is an increasingly prevalent and morbid cancer worldwide. Management of this cancer has changed significantly in the last decade through improved risk stratification and new therapies offering patients with locally advanced and metastatic disease more effective, less toxic, and more durable treatment options. Ongoing clinical trials are assessing new therapeutic options as well as optimizing existing regimens in efforts to better manage this cancer. The recent developments highlight the need for multidisciplinary care, especially for those with locally advanced and metastatic disease.

Abstract

cSCC is increasing in prevalence due to increased lifespans and improvements in survival for conditions that increase the risk of cSCC. The absolute mortality of cSCC exceeds melanoma in the United States and approaches that of melanoma worldwide. This review presents significant changes in the management of cSCC, focusing on improvements in risk stratification, new treatment options, optimization of existing treatments, and prevention strategies. One major breakthrough in cSCC treatment is the advent of immune checkpoint inhibitors (ICIs) targeting programmed cell death protein 1 (PD-1) and programmed death-ligand 1 (PD-L1), which have ushered in a renaissance in the treatment of patients with locally advanced and metastatic disease. These agents have offered patients with advanced disease decreased therapeutic toxicity compared to traditional chemotherapy agents, a more durable response after discontinuation, and improved survival. cSCC is an active field of research, and this review will highlight some of the novel and more developed clinical trials that are likely to impact cSCC management in the near future.

1. Introduction

cSCC consists of a wide range of clinical presentations, from low-risk squamous cell carcinoma in situ (cSCCis) to high-risk, locally advanced or metastatic tumors. The risk of metastatic disease in a patient with cSCC is approximately 2–4%, most commonly to the regional lymph nodes; however, the overall risk depends on the tumor subtype, location, and patient comorbidities [1,2]. It was not until 2010 that the American Joint Committee on Cancer (AJCC) separated cSCC into an independent staging system [3]. Improved risk stratification, particularly for patients without metastatic disease, has been an active area of research culminating in staging revisions and validation of staging criteria.
Shifts in patient demographics are influencing the incidence and development of cSCC. Patients with a history of drug-induced immunosuppression, HIV, lymphoma, leukemia, certain genetic syndromes (e.g., xeroderma pigmentosum), and solid organ transplant recipients (SOTR) are at higher risk of developing cSCC with a more aggressive clinical course [4,5,6,7,8,9,10]. SOTR remains the most significant acquired risk factor, increasing the risk of developing cSCC 5- to 113-fold, as well as the risk of developing local recurrence, metastasis, and overall mortality [11]. Owing to advances in anti-rejection therapy, donor-recipient matching, antimicrobial therapy, and transplant management over the last 30 years, SOTR patients are more likely to survive transplantation and live longer; the mean survival for a patient with a renal transplant is 22.79 years [12,13]. Similarly, patients with newly diagnosed HIV on antiretroviral therapy may have lifespans similar to that of the general population [14]. The significant gains in longevity for patients with these comorbidities will further increase the incidence of cSCC.
Identifying trends in cSCC epidemiology is challenging due to a multifactorial etiology coupled with inconsistent registry tracking—currently the CDC’s National Cancer Registry does not track cSCC. The annual incidence of cSCC is estimated to be 1.8 million cases [15]. For comparison, the combined annual incidence of other cancers in the United States is estimated to be 1.9 million; however, this figure is based on cancer registries that exclude non-melanoma skin cancer (NMSC) [16]. Worldwide, there has been a rapid increase in the incidence of cSCC due, in part, to the aforementioned demographic shifts as well as increased lifespan [17,18,19,20]. Several studies suggest a more rapid rise in cSCC incidence relative to basal cell carcinoma (BCC), which is currently the most common cancer in the United States [15,18,19]. Previous studies suggested a 4:1 incidence of BCC to cSCC, but recent data suggest a ratio of 1.69:1 overall [21]. Age is a particularly important risk factor for cSCC, with a BCC to cSCC ratio of 9.63:1 for patients aged 18–39 years and 1.33:1 for patients 65 years and older [21].
Unlike BCC, cSCC typically portends a worse prognosis. It is estimated that over 15,000 people die of cSCC in the United States annually [22]. Despite melanoma having a more aggressive clinical course than cSCC, the incidence of melanoma is significantly lower, resulting in approximately half the number of deaths in the United States [23]. However, globally, melanoma results in more deaths compared to cSCC, 62,800 versus 56,100, and disability-adjusted life years, 1.7 million versus 1.2 million [24].

2. Determining High-Risk cSCC and Prognostic Indicators

The 2022 National Comprehensive Cancer Network (NCCN) guidelines define high-risk and very high-risk cSCC based on the presence of risk factors for local recurrence, metastasis, or death [25]. In contrast to staging systems intended to stratify patients based on risk of metastasis, the NCCN guidelines intend to direct patient management. Risk factors for the general population are listed in Table 1. In addition, total number of tumors and frequency of development are considered risk factors in high-risk groups (i.e., immunosuppression or genetic syndromes predisposing cSCC). Based on a review of older NCCN guidelines, which used a similar approach to risk stratification as the current guidelines, 87% (n = 231) of cSCC diagnosed at a single institution were considered high-risk based on then-current guidelines, suggesting the guidelines are highly sensitive [26]. While the 2022 NCCN guidelines contain some modifications, the high-risk category is still highly inclusive and, therefore, less useful alone at predicting risk for metastasis.

2.1. Staging Systems

Despite the overall low risk of metastasis, cSCC is the second most common cancer worldwide, and the absolute number of patients with metastatic cSCC is increasing. Metastatic cSCC is associated with decreased survival compared to local disease, leading to attempts to stratify patients based on risk for metastasis [1,2,27]. Though clinical and dermatoscopic features aid in recognizing cSCC, histopathologic examination is the gold standard in the diagnosis of cSCC and identifies high-risk features, including tumor thickness, differentiation, and invasion into fat and nerves. Radiologic imaging is the diagnostic modality of choice to assess for bone invasion or metastatic disease in patients with high-risk cSCC.
Several staging systems have been proposed to predict risk of metastasis in cSCC (Table 2). The 7th edition AJCC staging manual first included a cSCC-specific staging system classifying tumors by size and additional risk factors including depth, degree of differentiation, anatomic location, and invasion of nerves and underlying skeletal structures [3,28]. More recent systems developed by a group from Brigham and Women’s Hospital (BWH) and Brueninger et al., summarized in Table 2, attempted to improve upon AJCC7 [29,30]. With the release of AJCC8 In 2017, some features from BWH were included, but only consisted of staging criteria for cSCC of the head and neck [31]. The BWH and AJCC8 systems are the most commonly used staging systems in the United States.
Schmitt et al. conducted a meta-analysis comparing the performance of AJCC7 to BWH in predicting sentinel lymph node biopsy (SLNB) positivity [32]. The rate of positivity in the BWH system was 7.1% for T2a, 29.4% for T2b, and 50.0% for stage T3 compared to the AJCC7′s 11.2 %% for T2 and 60% for T4 [32]. Both systems had 0 positive cases in the T1 stage [32]. None of the cases were categorized as AJCC7 stage T3. The authors concluded that the BWH system more accurately predicted SLNB positivity.
Ruiz et al. compared AJCC8 performance with BWH when applied to a large cohort at a single center institution, focusing on metastases and deaths [33]. The high stage AJCC8 (T3/T4) and BWH (T2b/T3) patients had indistinguishable outcomes; however, the BWH high stage subset was half the size as the AJCC8, 63 versus 121 patients, respectively [33]. The BWH system had a higher specificity and positive predictive value compared to AJCC8 [33]. Venables et al. compared AJCC8 and BWH in a larger cohort from the National Disease Registration service in the UK and found similar results, noting that BWH has a higher specificity and positive predictive value compared to AJCC8, whereas AJCC8 has a slightly higher negative predictive value [34]. Overall, the data suggest that AJCC8 upstages low-risk disease, and BWH outperforms AJCC8 in identifying low-risk cSCC.

2.2. Sentinel Lymph Node Biopsy

Given the predilection of cSCC for regional lymph nodes, SLNB is well suited to detect occult metastatic disease. The overall rate of SLNB positivity is 13.9% and false negativity is 4.6% [35]. Patients with a positive SLN have a higher risk of recurrence, further metastasis, and death compared to SLN negative patients despite treatment with neck dissection and/or adjuvant radiation therapy (RT) [36,37]. Thus, SLNB adds prognostic value in high stage tumors but does not appear to improve patient outcomes based on the limited evidence available. The role of SLNB in management of cSCC remains unclear and further studies are ongoing.

2.3. Gene Expression Profiling and Tumor Biomarkers

A recently developed prognostic 40-gene expression profile test performed on biopsied cSCC tissue allows patients to be stratified into three groups based on three-year metastasis risk [38]. A retrospective analysis of 300 patients assayed determined the risk of metastasis was 9% for class 1 (low risk), 21% for 2A (high risk), and 63% for 2B (highest risk) [39]. Management recommendations have been proposed for the different risk classes in combination with the staging systems; however, gene expression profiling has not been incorporated into the NCCN or American Academy of Dermatology (AAD) guidelines [25,40].
Expression of individual tumor markers is prognostic in cSCC. Tumor expression of programmed death-1 ligand (PD-L1) carries increased risk of nodal metastasis according to a meta-analysis of seven studies (odds ratio 2.34) [41]. Additional tumor markers associated with local recurrence, perineural invasion (PNI), metastasis, and poor survival include inositol polyphosphate 5-phosphatase, p300, telomerase reverse transcriptase, CD133, thymidine kinase 1, Myb-related protein B, and long noncoding RNAs [42,43,44,45,46,47]. Further studies are needed to determine appropriate testing protocols for tumor markers in patients at risk for advanced cSCC and their implications.

2.4. Imaging

Staging imaging studies are not indicated for all cases of cSCC due to the low overall risk for metastasis. Imaging is most commonly employed to screen for subclinical regional metastasis. Although limited data are available regarding imaging of cSCC, it is recommended that imaging of the regional lymph node basin be considered for BWH stage T2b and T3 cSCCs [48,49,50].
In the United States, nodal basins are typically evaluated by computed tomography (CT) with contrast due to the relatively low cost, high speed, ability to assess cortical bone involvement, and spatial information provided [51]. Ultrasound (US) is another option that is 91% sensitive in detecting nodal disease in head and neck cSCC [52]. Advantages to US include avoidance of ionizing radiation and contrast, as well as potential concomitant fine needle aspiration if a concerning node is identified. However, reliable US results may depend on technician experience and facility case volume. Positron emission tomography (PET) and magnetic resonance imaging (MRI) are less frequently used for nodal basin assessment, but the latter is well suited for assessing PNI [51].
Imaging may be considered for locally advanced disease or pre-operative planning purposes [53]. Furthermore, there may be benefits to post-treatment surveillance imaging. Baseline and surveillance imaging identified nodal disease not detected on clinical exam in 21% of patients in a cohort of high-risk cSCCs [54]. There are no consensus guidelines regarding surveillance imaging, however over 75% of tumor recurrences and metastases occur within 2 years [55,56]. In addition to a clinical exam with nodal assessment, surveillance imaging with either CT or US can be considered every 4–6 months for 2 years following definitive treatment [48].

3. Treatment

3.1. Topical Treatment

Surgical excision is the gold standard treatment for cSCC, but topical therapies are often employed for lower risk cSCCis and may be utilized as an adjunct in non-surgical candidates or those who refuse surgery. The most frequently used agents are 5-fluorouracil (5-FU), imiquimod, and destructive acid (e.g., trichloroacetic acid). There are limited quality data regarding efficacy, but Bennardo et al. conducted a systematic review encompassing 49 patients treated with 5% 5-FU, 3.75–5% imiquimod, 0.1% tazarotene, and/or 80% trichloroacetic acid [57]. The response rate was 95% when topical modalities were used in combination (5-FU + imiquimod, carbon dioxide laser + 5-FU, intralesional 5-FU + topical trichloroacetic acid) and dropped to 67% for monotherapy [57]. The majority of treatment failures were in the cohort treated with tazarotene [57].
In the author’s experience, adjunctive topical therapy with either 5-FU or imiquimod is a valuable tool to decrease surgical mobidity and disfigurement, particularly for Mohs cases, in which there is residual cSCCis on a peripheral margin after clearing the invasive tumor, or when operating in a patient with field cancerization. Topical therapies are well-suited for human papillomavirus (HPV)-associated cSCCis that are multifocal (i.e., bowenoid papulosis of the genitals) and have been reported as an adjunct when surgical margins are positive in HPV-associated cSCC [58].

3.2. Destruction

Destruction with liquid nitrogen or curettage and electrodesiccation (C&E) are treatment options for low-risk cSCC. A pooled analysis of eight retrospective observational studies revealed a recurrence rate of 1.7% and 0.8% for C&E and cryotherapy, respectively [59]. However, most of the lesions in this analysis were smaller, lower risk cSCC. The recurrence rate of cSCC greater than 2 cm treated with C&E was 11.8% [59]. A disadvantage of destruction, especially on the head and neck, is cosmesis. Seventeen percent of patients were deemed to have a “poor” cosmetic outcome in one study, and 54% had a satisfactory cosmetic outcome [59].

3.3. Surgical Excision

Surgical excision is effective for most cSCCs. Both NCCN and AAD recommend 4- to 6-mm clinical margins for standard excision of low-risk cSCC, with the AAD specifically recommending excision to the depth of the subcutaneous adipose tissue [25,40].

3.4. Mohs Micrographic Surgery

Mohs micrographic surgery (MMS) results in superior histopathologic verification of complete tumor extirpation and allows for maximum conservation of tissue compared to standard surgical excision [59]. This technique is especially important for cSCCs located in high-risk anatomic sites, including those on the head and neck. Rowe et al. conducted a systematic review, demonstrating a 5-year local recurrence rate of 3.1% for primary cSCC treated with MMS [27]. For recurrent cSCC, the 5-year recurrence rate after MMS was 10.0% versus 23.3% for standard excision [27]. The findings are summarized in Table 3. No RCTs or prospective cohort studies comparing MMS to other treatment modalities for cSCC have been published.

3.5. Radiation Therapy

RT is an option for primary treatment of cSCC when surgery is contraindicated, associated with high morbidity, or not preferred by the patient. Because of potential long-term sequelae, patients over the age of 60 years old are preferred candidates. High-level evidence of this treatment modality is lacking; current data are limited by small patient numbers, variable follow-up durations, and heterogeneity of RT modality employed. Cure rates of RT have been shown to be lower than standard surgical excision [59]. Smaller and thinner tumors may be more responsive to radiation therapy. Other disadvantages of RT include lack of margin control, prolonged course of therapy, post-radiation skin changes (chronic dermatitis and fibrosis), and potential increased risk of future cancers in the treatment field. RT as an adjuvant modality is often employed following surgical treatment of high-risk SCC and will be discussed later [60,61].

3.6. Photodynamic Therapy

Photodynamic therapy (PDT) is widely used for field treatment actinic keratoses and involves topical application of a photosensitizer, such as aminolevulinic acid or methyl aminolevulinate. Due to reports of high recurrence with PDT, as well as a lack of high-level evidence for its use, PDT is generally not recommended for cSCC [59,62,63]. PDT is better suited for cSCCis, which is confined to the epidermis and within the therapeutic range of PDT. A PDT cure rate for cSCCis of 86–93% can be achieved, with a 2-year sustained clearance rate of 68–71% [62].

3.7. Laser

Limited data exist for laser treatment of cSCC in the literature [59]. No study has shown that laser has efficacy in treating cSCC; however, case series do outline successful treatment of cSCCis with ablative laser therapy [64]. In the largest case series, 44 patients with 48 cSCCis treated with one or more passes of carbon dioxide laser at 2 W/cm2 demonstrated a 6.8% recurrence rate at 18 months mean follow-up [65].
Laser-assisted drug delivery augments the distribution and penetration of topically applied treatments by utilizing ablative fractional laser (AFL). Two randomized controlled trials showed clearance of cSCCis with AFL-PDT was 87.0–87.5% versus 50.0–55.3% for the PDT-only arm at 12 months [66,67]. A case series of 16 patients demonstrated 100% clearance of cSCCis at 8 weeks after a single treatment of AFL followed by topical 5% 5-FU under occlusion for 7 days [68].

3.8. Cytotoxic Chemotherapy

Locally advanced or metastatic cSCCs not amenable to surgical excision or radiation therapy require a more aggressive therapeutic approach [69]. Prior to the advent of immunotherapy, cytotoxic chemotherapies such as carboplatin, cisplatin, bleomycin, and 5-FU were the mainstay of advanced cSCC treatment, either as a monotherapy or combination therapy [69,70,71]. Most clinical data supporting these treatment regimens are limited single-arm studies and case series with response rates ranging from 17% to 84% [69,72,73,74]. Combination therapies demonstrate higher efficacy than single agent therapy; however, there is a lack of durable response following cessation of treatment [69]. In many studies, chemotherapies were combined with other treatment modalities such as RT or surgery, which further confounds long-term data analysis of these treatment regimens [75]. Cytotoxic agents demonstrate short progression free survival (PFS) and/or overall survival (OS) [74]. In cases with longer tumor remission, patients received subsequent surgery or RT [75].
Treatment-related toxicities are a significant concern for patients undergoing cytotoxic chemotherapy [69]. More than one-third of patients treated with combination therapy experienced grade 3 to 4 hematologic adverse events, including anemia and neutropenia, in available studies [71,76]. Cytotoxic agents are not FDA-approved for the treatment of advanced cSCC and are not preferred due to substantial treatment-associated morbidity and lack of durable response [70]. Due to a lack of higher-powered randomized controlled trials analyzing cytotoxic chemotherapy regimens, no formal treatment guidelines have been established.

3.9. Immunotherapy

Intralesional interferon represented the first use of immunotherapy for the treatment of cSCC in 1992 [77,78]. Since then, there has been an increased understanding of the immune system’s role in cSCC development, surveillance, and targeting of cancerous cells for elimination [69]. Chronic immunosuppression significantly increases the incidence of NMSC through impaired immunosurveillance.
Repeated skin exposure to mutagens such as ultraviolet radiation is a major pathogenic factor in the development of cSCC [79]. Tumor cells escape immunosurveillance by activation of regulatory checkpoints that place a physiologic brake on the immune system [80]. Immunotherapy with ICIs promotes T-cell mediated tumor destruction through the removal of the brakes on the immune system [69]. Highly mutated tumors express more tumor-associated neoantigens and are more likely to respond to ICI therapy [81,82,83]. cSCC has a relatively high number of mutations compared to other solid tumors, making it well suited for ICI therapy [69,81,82].

3.9.1. Program Death 1 Inhibition

PD-1 is a transmembrane receptor expressed on T-cells, B-cells, and natural killer cells and serves as an immune regulating checkpoint. PD-L1, its ligand, is expressed on tumor and antigen-presenting cells [84]. The binding of PD-L1 to the PD-1 receptor leads to peripheral T-cell exhaustion and immunosuppression in the local tumor microenvironment [84,85]. In 2018, the introduction of immunotherapy with anti-PD-1 antibodies resulted in a paradigm shift in the treatment of advanced cSCC, offering increased efficacy, improved safety, and durability after treatment discontinuation [69].
Cemiplimab is a human monoclonal immunoglobulin G4 antibody directed against PD-1 [86]. In 2018, it became the first FDA-approved treatment for locally advanced or metastatic cSCC. It is approved in the European Union for the same indications. Approval was driven by two clinical trials involving 137 patients with advanced and/or metastatic cSCC [87,88]. In the phase I and II trials, patients received intravenous cemiplimab at a dose of 3 mg/kg every 2 weeks for up to 48 weeks (phase 1), 96 weeks (phase 2), or until there was disease progression or unacceptable toxicity [86]. The combined phase I and II populations had an objective response rate (ORR) of 47% [86,89]. Sixty-five percent of patients had durable disease control, defined as greater than 105 days without disease progression, and 54% of patients achieved disease control of at least 6 months in the phase I cohort [86]. Sixty-one percent of patients with metastatic cSCC in the Phase II study achieved durable disease control, with 57% achieving disease control for at least 6 months [86]. The majority of adverse reactions included grade 1 or 2 reactions such as fatigue, rash, nausea, diarrhea, and constipation [86,90,91]. Grade 3 or higher adverse reactions occurred less frequently, and included hypo- and hyperthyroidism, and type 1 diabetes mellitus, pneumonitis, colitis, hepatitis, nephritis, hypertension, adrenal insufficiency, and anemia [86,90,91].
A 43-month follow-up analysis of the phase II study (summarized in Table 4) showed a stable ORR of 47.2%, with improvement seen in the cohort with metastatic cSCC, which had an ORR increase from 42.9% to 46.4% [92]. Two additional patients had a complete response (CR). The improvements in ORR and CR demonstrate the findings of greater response with longer follow-up. Duration of response continued to improve across all cohorts, and the safety profile remained similar to prior studies with anti PD-1 agents [86,90,91].
Pembrolizumab is a humanized monoclonal antibody that binds the PD-1 receptor on T-cells that received FDA approval in 2020 for the treatment of recurrent or metastatic cSCC not amenable to surgery or RT. Approval stemmed from the KEYNOTE-629 Phase II clinical trial of 105 patients with recurrent or metastatic cSCC with a median follow-up of 11.4 months [93]. Eighty-six percent of patients had previously been treated with at least one systemic therapy prior to the trial. Pembrolizumab was administered at a dose of 200 mg every 3 weeks for a maximum of 24 months or until disease progression or toxicity. The combined recurrent and metastatic cSCC cohort had an ORR of 34.3%. Most adverse reactions were grade 1 or 2 and included diarrhea, fatigue, pruritus, and constipation. Grade 3 or higher adverse reactions occurred frequently and included colitis, hepatitis, endocrinopathies, pneumonitis, nephritis, and skin toxicity.
Hughes et al. subsequently analyzed the KEYNOTE-629 trial (summarized in Table 5) at a mean follow-up of 27.2 months and a new cohort of 54 patients with locally advanced cSCC treated with pembrolizumab at a mean follow-up of 14.9 months [94]. The ORR increased slightly to 35.2% in the recurrent and metastatic cohort, and the locally advanced cohort had an ORR of 50.0%. Of note, efficacy comparisons across trials are difficult to interpret, particularly because locally advanced cSCC in the KEYNOTE-629 clinical trial did not include recurrent disease, while locally advanced cSCC in the cemiplimab trials included recurrent disease [93,94]. Eleven percent of patients experienced grade 3 to 5 treatment related adverse reactions, and 8% of patients had grade 3 to 5 immune related adverse reactions [94]. Overall, a durable response and robust anti-tumor response were observed in both cohorts with an acceptable safety profile.

3.9.2. Cytotoxic T-Lymphocyte-Associated Antigen 4 Inhibition

Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) is an immune checkpoint receptor expressed on T-cells. The binding of CTLA-4 to B7, its costimulatory protein receptor on antigen presenting cells, decreases T-cell regulatory activation [85,95]. Blockage of this pathway increases anti-tumor response through T-cell activation [83].
Ipilimumab is a human monoclonal antibody against CTLA-4 and is FDA-approved for treatment of unresectable or metastatic melanoma [96]. While groundbreaking for the treatment of melanoma, ipilimumab has a much smaller role for cSCC. Day et al. reported a case of a patient with both metastatic melanoma and metastatic cSCC treated with ipilimumab monotherapy for 4 cycles [97]. The patient demonstrated clinical benefit and a durable response of all tumors with grade 2 or less adverse events [97]. Larger randomized controlled trials are necessary to assess anti-CTLA-4 antibodies for cSCC treatment. Given the side effect profile of ipilimumab, other ICIs are typically favored.

3.10. Targeted Therapy

The epidermal growth factor receptor (EGFR) is a tyrosine kinases receptor involved in signaling pathways critical for cell proliferation, migration, differentiation, regulation, and survival [98]. Tumor development often involves abnormal activation of the EGFR pathway, and up to 80% of cSCC and 100% of metastatic cSCC overexpress EGFR [99,100]. Commercially available EGFR inhibitors include the oral small molecule tyrosine kinase inhibitors, erlotinib and gefitinib, and intravenous monoclonal antibodies, cetuximab and panitumumab [101]. Although not considered first line systemic therapy, EGFR inhibitors are considered in patients with contraindications to ICIs or ineligible for clinical trials. EGFR inhibitors may be used alone or in combination with conventional chemotherapy or RT [25].
Data supporting the efficacy of EGFR inhibitors in the treatment of cSCC are limited to a few prospective studies that are summarized in Table 6. Cetuximab monotherapy showed an ORR of 28%, including a CR in 6% and partial response (PR) in 22%, when used for at least 6 weeks in a phase II clinical study [100]. PFS and OS were 4.1 months and 8.1 months, respectively [100]. Eighty-seven percent of patients experienced a grade 1 or 2 acneiform eruption [100]. Sixty-one of patients experienced a grade 3 and 4 toxicity, including infection, bleeding of the tumor, and infusion reaction [100]. A small, 16-patient study of panitumumab monotherapy demonstrated an ORR of 31%, including CR in 13% and PR in 19% [102]. PFS and OS were 8 months and 11 months, respectively. Notably, all patients included in the study were being treated as second line therapy. Five of the sixteen patients experienced grade 3 or 4 toxicities, including four with skin toxicity [102]. Oral gefitinib demonstrated an ORR of 16%, all of which were PR, in a phase II study of 37 patients with cSCC [103]. PFS and median OS were 3.8 months and 12.9 months, respectively [103]. Oral erlotinib demonstrated an ORR of 10%, all of which were PR, in a phase II trial of 29 patients with cSCC [104]. PFS and OS were 4.7 months and 13 months, respectively [104]. Treatment with these agents may be limited by shorter duration of response and progression free survival compared to more modern systemic therapies.
Side effects of EGFR inhibitors are typically less severe than those seen with cytotoxic chemotherapy. The most common adverse event is an acneiform eruption, reported in 50–100% of patients and a major cause of drug cessation or poor treatment adherence [105]. Interestingly, the occurrence and severity of the acneiform eruption is positively correlated with tumor response [100,106]. Patients who developed this eruption secondary to EGFR inhibitors have an increased PFS and a tendency for a longer OS [100]. Diarrhea is also a common adverse effect of EGFR inhibitors due to the expression of EGFR in epithelial cells of the gastrointestinal tract, with incidences ranging from 27% to 87% in clinical trials for a variety of malignancies [107]. Other reported side effects include fatigue, rash, malaise, infection, neutropenia, peripheral sensory neuropathy, weight loss, and elevated liver transaminases [48]. Despite these adverse effects, EGFR inhibitors tend to be better tolerated by patients compared to cytotoxic chemotherapies.

3.11. Intratumoral Injection

cSCC is most commonly limited to the skin, allowing direct access of therapeutics to the tumor site. While systemic agents can elicit a variety of adverse events, intratumoral injection increases local drug concentrations and potentially diminishes systemic toxicities [108]. Intralesional treatment may be preferred in poor surgical candidates or those with challenging tumor location or high tumor burden. Intratumoral injections are user-dependent, and correct injection technique is crucial to the successful delivery of a drug.

3.11.1. Methotrexate

Methotrexate (MTX) is an anti-metabolite therapy commonly used for its anti-tumoral and immunosuppressive effect against tumors and autoimmune disease [109]. As a competitive inhibitor of dihydrofolate reductase, it prevents the conversion of dihydrofolate to tetrahydrofolate, a step crucial to de novo purine and pyrimidine synthesis [110]. Classically, systemic MTX was used intravenously for a variety of malignancies, including childhood leukemia [111]. More recently, methotrexate has been used intralesionally in easily accessible tumors, particularly cSCC.
Intralesional methotrexate has had some success in the treatment of localized cutaneous squamous cell carcinoma without metastasis. In a single-center, prospective study that included 65 cases of cSCC treated with intralesional MTX prior to surgical treatment, 92.3% of patients showed a clinical response, with 58.5% of patients having no residual dysplasia on the subsequent excision pathology [112]. The mean tumor surface area of clinical responders was 1.69 cm2 versus 7.72 cm2 in the non-responder group, and all tumors were treated with the same protocol, two 20 mg treatments spaced 1 week apart, regardless of the tumor size [112]. Annest et al. treated 38 keratoacanthoma-type cSCC with intralesional MTX and observed a cure rate of 92% [113]. On average, two injections were given, 18 days apart, with an injection volume of 1 mL and 17.6 mg/mL of MTX [113]. MTX is metabolized in the liver and renally excreted. Although injection is intralesional, systemic adverse events have been reported [113]. MTX toxicity is characterized by nausea, vomiting, diarrhea, pancytopenia, liver dysfunction, acute renal failure, pulmonary symptoms, stomatitis, mucositis, and gastrointestinal or cutaneous ulcerations [114].

3.11.2. 5-Fluorouracil

5-FU is fluoropyrimidine that is converted to its active metabolite in the cell and disrupts RNA synthesis [115]. Intralesional 5-FU has shown success in the treatment of keratoacanthomas. In 1978, Odom et al. treated 14 patients with 26 keratoacanthomas using weekly intralesional 5-FU [116]. All but one lesion cleared with an average of 2.8 injections, and of the 13 patients observed, none had recurrence at 12 months. Intralesional 5-FU has shown efficacy in patients with large keratoacanthomas, recurrent keratoacanthomas, and invasive SCC [117]. Larger-scale randomized controlled trials are needed to better characterize the use of intralesional 5-FU for the treatment of cSCC. In general, 5-FU toxicities include fever, mucositis, nausea, vomiting, diarrhea, and myelotoxicity [118].

3.12. Combination Therapy and Adjuncts

While surgical excision of cSCC is often the standard of care, combining therapeutic modalities has been common practice since for high-risk cSCC. Systemic therapy can be coupled with surgery or RT as adjuvant or neoadjuvant therapy [119].
Adjuvant RT reduces the likelihood of recurrence and metastasis after surgical excision [53]. However, the majority of studies on adjuvant RT have been retrospective in design [119]. A retrospective study by Wang et al. showed a higher survival rate for patients with cSCC and cervical lymph node involvement treated with surgery plus adjuvant RT, compared to those treated with surgery alone [120]. Another study by Trosman et al. showed that adjuvant RT and surgery did not significantly improve 2-year disease free survival rates compared to surgery alone [121]. These contradictions continue in numerous studies investigating adjuvant RT, and there is little evidence to help determine which high-risk cSCC tumors would benefit from adjuvant RT. AAD guidelines recommend adjuvant RT for primary cSCC with concerning PNI or otherwise high risk for regional or distant metastasis, following surgical treatment [40]. However, the guidelines do note that there is no high-level evidence about the effectiveness of this approach [40]. The NCCN recommends adjuvant RT for any cSCC with extensive perineural involvement, large nerve involvement, or in which tissue margins remain positive after definitive surgery [25].
Adjuvant chemotherapy includes the use of various antitumor agents including platinum-based agents (e.g., cisplatin and carboplatin) as well as anti-metabolites (e.g., 5-FU). Similar to adjuvant RT, studies on adjuvant chemotherapy are predominantly retrospective in nature with small patient cohorts [53]. Due to the significant toxicities of chemotherapy, these agents are not frequently used in patients with significant comorbidities [122]. Future prospective trials are necessary to optimize which locally advanced and metastatic tumors would benefit from adjuvant chemotherapy, adjuvant RT, or a combination of the two.
For locally advanced cSCC, consideration of immunotherapy and/or RT is recommended when surgery is unable to clear the tumor or would result in significant disfigurement or loss of function [25]. Numerous trials are investigating a combination of targeted therapies and immunotherapies in combination with chemotherapy or RT in an adjuvant and neoadjuvant setting [53]. At this time, due to a lack of prospective randomized controlled studies, a multidisciplinary approach and consideration of referral to a cancer center is recommended for discussion to optimally combine these treatment modalities for patients with locally advanced and/or metastatic cSCC.

4. Chemoprevention

Oral retinoids have been shown to reduce the incidence of cSCC in patients with certain high-risk factors—SOTR, genetic predisposition (e.g., xeroderma pigmentosum), and prior PUVA exposure; however, the retinoids do not produce a durable response and benefits quickly abate after cessation of therapy [123,124,125,126,127,128,129]. The benefit is more pronounced in SOTR patients with a prior history of cSCC, compared to SOTR patients without [125,127]. Topical retinoids and oral isotretinoin have failed to show benefits in preventing cSCC development [130,131,132]. There are conflicting data regarding the benefits of oral retinol [132,133]. Patients on oral retinoid therapy require regular monitoring given the risks, which include teratogenicity, hepatitis, hyperlipidemia, alopecia, and mucocutaneous dryness. Based on the limited data, the AAD recommends against the use of retinoids for chemoprevention, with the sole exception that acitretin may be used in patients with a history of SOTR and cSCC [40]. The NCCN acknowledges that oral retinoids may be effective in reducing the development of cSCC in some high-risk patients other than SOTR [25].
The benefits of chemoprevention are less distinct for other therapies and patient groups at high risk. Weinstock et al. demonstrated a 75% reduced risk of developing cSCC at 12 month follow-up with the prophylactic use of 5-FU cream compared to placebo in patients with a history of NMSC [134]. Study patients were included if they had a history of two keratinocyte tumors in the last 5 years, one of which was located on the head or neck. Patients with a history of SOTR, genetic predisposition, PUVA, CTCL, or prior radiation were specifically excluded from the study.
Nicotinamide, a variant of vitamin B-3, has shown efficacy in the chemoprevention of actinic keratoses and NMSC in those at elevated risk. In a phase 3 double-blinded randomized controlled trial, nicotinamide 500 mg BID resulted in a 30% reduction in cSCC after 12 months of therapy [135]. Smaller reductions were found in the incidence of basal cell carcinoma (20%) and actinic keratoses (13%). Patients were included if they had a history of 2 NMSC in the past 5 years, and those with immunosuppression or genetic skin cancer syndromes were excluded. Similar to oral retinoids, the chemopreventive benefit did not persist significantly after discontinuation of therapy. A meta-analysis of the use of nicotinamide in preventing cSCC, which included patients with a history of SOTR, found an overall rate ratio of 0.48 (0.26–0.88, 95% CI) further supporting its use as a chemopreventive agent [136]. Unlike a related drug, niacin, which often causes flushing and other vasodilatory phenomena, nicotinamide is well tolerated. There was no significant difference in adverse events between the treatment and placebo arm. The AAD guidelines recommend against the use of nicotinamide for chemoprevention; however, these guidelines were last updated in 2018 and based on the limited evidence available at the time [40]. The NCCN guidelines, most recently published in May 2022, acknowledge that nicotinamide may reduce the development of cSCC, but do not recommend for or against its use [25].
Capecitabine is an oral precursor to 5-FU, an agent commonly used as a topical treatment of NMSC and actinic keratoses. Lewis et al. first reported capecitabine-associated inflammation of actinic keratoses in a patient treated for metastatic colorectal carcinoma [137]. This and other subsequent reports led to studies evaluating the use of capecitabine as a chemopreventive agent. In a systematic review, 13 of 18 SOTR patients demonstrated at least a 50% reduction in cSCC during 12 months of therapy with capecitabine [138]. Fifty-six percent of all patients included in the review discontinued therapy due to disease progression or adverse events, including an unrelated death in one patient [138]. Interestingly, capecitabine may also reduce the incidence of basal cell carcinoma in patients with a history of SOTR [139]. Larger studies are needed to better define the chemopreventive benefits. Common adverse effects included fatigue, hand-foot syndrome, diarrhea, nausea/emesis, mucositis, anemia, and hyperuricemia/gout. Further investigation is necessary to better determine the risks and benefits of capecitabine in the prevention of cSCC; however, it may be considered in patients with very high rates of cSCC formation and field cancerization who have failed other treatment strategies.
Despite early evidence that NSAIDs may decrease the risk of cSCC, a subsequent meta-analysis showed no chemopreventive benefit [140]. Both the NCCN and AAD advise against the use of NSAIDs for chemoprevention of cSCC due to the lack of efficacy and increased risk of cardiovascular and gastrointestinal adverse events [25,40].
PDT is widely used for field treatment of actinic keratoses and has been used off-label for multiple indications, including the treatment of low-risk NMSC, as discussed earlier. Data on the use of PDT for prevention of NMSC are limited. One study of 12 SOTR patients found a 79% and 95% reduction in the number of cSCC at 12 and 24 months, respectively, when treated with cyclic PDT every 4–8 weeks [141]. Adverse events were mild and included erythema, edema, desquamation, and crusting. There were significant limitations to the study, including a short observation period and a high number of cSCCs treated at baseline immediately prior to PDT, possibly overstating the benefits of the treatment. Further investigation of this relatively low-risk preventative modality is warranted.

5. Future Directions

As of May 2022, there are 179 clinical trials for cSCC. The biggest trend with the current trials is the use of ICIs, including three novel agents targeting PD-1 and/or PD-L1 [142,143,144]. Many trials are investigating the efficacy of neoadjuvant, adjuvant, or combined neoadjuvant and adjuvant ICI therapy with surgery and/or RT. Several novel agents are being combined with existing ICI therapy, specifically medications stimulating IL-2, IL-7, IL-15, TLR-7/8, TLR-9, or inhibiting c5a and EGFR/TGFβ [145,146,147,148,149,150,151,152]. In an effort to decrease systemic exposure and risk of ICIs, one trial is evaluating low dose intratumoral injection of cemiplimab [153].
Investigational drugs targeting CD-47, CD-40, TGFβ1, COX-2, and the STING pathway are being studied as monotherapy for cSCC [154,155,156]. Several unique types of therapy delivery are also being studied. There are two trials evaluating photoimmunotherapy with ICIs [157,158]. Photoimmunotherapy involves systemic infusion of a medication followed by local activation with a specific wavelength of light. NCT05377905 utilizes a microneedle array that delivers intratumoral doxorubicin; notably, patients with a history of SOTR are included in this study [159]. Several studies are evaluating specific applications of RT, including the use of diffuse alpha-emitter RT, which involves intratumoral placement of radium-224 and short-range emission of alpha particles, potentially limiting collateral tissue damage [160]. IFX-Hu2.0 is a DNA plasmid that induces tumoral expression of streptococcal antigen, potentially increasing immunosurveillance, and the plasmid vehicle potentially avoids side effects associated with viral-mediated gene therapy [161,162].
Oncolytic viruses, viruses that have been genetically modified or selected for anti-tumoral activity, have been used previously in melanoma and other solid tumors. Talimogene laherparepvec (T-VEC) is a genetically modified herpes virus used in the treatment of stage III and IV melanoma and was the first to be FDA approved in the US. T-VEC is currently being studied in small trials for the treatment of cSCC as monotherapy or in combination with panitumumab or nivolumab [163,164,165]. TBio-6517, ONCR-177, RP-1, MEM-288, and Daromun are oncolytic viruses currently in phase 1 and 2 trials. They influence both tumor and tumor microenvironment with activity on Flt3, CTLA4, IL-2, IL-12, CCL4, PD-1, CM-CSF, TNF, and GALV-GP R-. TBio-6517 and RP-1 are the farthest along, with trials combining intratumoral injection of the oncolytic virus with systemic ICIs [166,167].
Treatment of locally advanced and metastatic cSCC in SOTR patients poses significant challenges. First-line treatments, including RT with chemotherapy or EGFR inhibitors, are limited by their modest efficacy and low tolerability. ICI treatment is limited by the risk of grafted organ rejection, which approaches 42% in renal transplants [168]. NCT04339062 is evaluating the use of cemiplimab in patients with a history of renal transplant, incorporating pre-infusion prednisone and sirolimus to reduce the risk of graft rejection [169]. Intratumoral RP-1 is also being investigated as monotherapy in patients with SOTR [170].
Given the variable and dynamic response of ICI therapy, several trials are assessing factors that may better predict response to therapy. Based on data from murine studies and patients with melanoma, two trials are evaluating fecal microbiota, including one that involves fecal transplant from ICI responders to ICI non-responders [171,172]. Two large cohort studies are investigating various factors and their potential association with overall survival, incidence of immune-related adverse events, and quality of life [173,174].

6. Conclusions

Traditional surgical and destructive methods remain the standard of care for low-risk cSCC; however, management of high-risk, locally advanced, and metastatic cSCC has evolved significantly in the last decade to include more efficacious, durable, and tolerable therapeutic options. With the increasing incidence of cSCC and growing number of patients with high risk factors, it is imperative that clinicians continue to expand their treatment armamentarium to include the most recent advances.

Author Contributions

Conceptualization, C.B.; writing—original draft preparation, C.B., S.K., Q.-B.N., N.A.-F. and K.M.W.; writing—review and editing, C.B., O.M.C., H.Q.D., S.S. and M.R.M.; supervision, M.R.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tokez, S.; Wakkee, M.; Kan, W.; Venables, Z.C.; Mooyaart, A.L.; Louwman, M.; Nijsten, T.; Hollestein, L.M. Cumulative incidence and disease-specific survival of metastatic cutaneous squamous cell carcinoma: A nationwide cancer registry study. J. Am. Acad. Dermatol. 2022, 86, 331–338. [Google Scholar] [CrossRef]
  2. Brantsch, K.D.; Meisner, C.; Schönfisch, B.; Trilling, B.; Wehner-Caroli, J.; Röcken, M.; Breuninger, H. Analysis of risk factors determining prognosis of cutaneous squamous-cell carcinoma: A prospective study. Lancet Oncol. 2008, 9, 713–720. [Google Scholar] [CrossRef]
  3. Edge, S.B. American Joint Committee on Cancer. In AJCC Cancer Staging Manual, 7th ed.; Springer: New York, NY, USA, 2010; ISBN 978-0-387-88440-0. [Google Scholar]
  4. Hortlund, M.; Arroyo Mühr, L.S.; Storm, H.; Engholm, G.; Dillner, J.; Bzhalava, D. Cancer risks after solid organ transplantation and after long-term dialysis. Int. J. Cancer 2017, 140, 1091–1101. [Google Scholar] [CrossRef]
  5. Manyam, B.V.; Garsa, A.A.; Chin, R.-I.; Reddy, C.A.; Gastman, B.; Thorstad, W.; Yom, S.S.; Nussenbaum, B.; Wang, S.J.; Vidimos, A.T.; et al. A multi-institutional comparison of outcomes of immunosuppressed and immunocompetent patients treated with surgery and radiation therapy for cutaneous squamous cell carcinoma of the head and neck. Cancer 2017, 123, 2054–2060. [Google Scholar] [CrossRef] [Green Version]
  6. Kraemer, K.H.; DiGiovanna, J.J. Forty years of research on xeroderma pigmentosum at the US National Institutes of Health. Photochem. Photobiol. 2015, 91, 452–459. [Google Scholar] [CrossRef] [Green Version]
  7. Miranda, M.B.; Lauseker, M.; Kraus, M.-P.; Proetel, U.; Hanfstein, B.; Fabarius, A.; Baerlocher, G.M.; Heim, D.; Hossfeld, D.K.; Kolb, H.-J.; et al. Secondary malignancies in chronic myeloid leukemia patients after imatinib-based treatment: Long-term observation in CML Study IV. Leukemia 2016, 30, 1255–1262. [Google Scholar] [CrossRef] [Green Version]
  8. Jensen, A.O.; Svaerke, C.; Farkas, D.; Pedersen, L.; Kragballe, K.; Sørensen, H.T. Skin cancer risk among solid organ recipients: A nationwide cohort study in Denmark. Acta Derm. Venereol. 2010, 90, 474–479. [Google Scholar] [CrossRef] [Green Version]
  9. Jensen, P.; Hansen, S.; Møller, B.; Leivestad, T.; Pfeffer, P.; Geiran, O.; Fauchald, P.; Simonsen, S. Skin cancer in kidney and heart transplant recipients and different long-term immunosuppressive therapy regimens. J. Am. Acad. Dermatol. 1999, 40, 177–186. [Google Scholar] [CrossRef]
  10. Patel, R.V.; Clark, L.N.; Lebwohl, M.; Weinberg, J.M. Treatments for psoriasis and the risk of malignancy. J. Am. Acad. Dermatol. 2009, 60, 1001–1017. [Google Scholar] [CrossRef]
  11. Berg, D.; Otley, C.C. Skin cancer in organ transplant recipients: Epidemiology, pathogenesis, and management. J. Am. Acad. Dermatol. 2002, 47, 1–17; quiz 18–20. [Google Scholar] [CrossRef] [PubMed]
  12. Rana, A.; Godfrey, E.L. Outcomes in Solid-Organ Transplantation: Success and Stagnation. Tex. Heart Inst. J. 2019, 46, 75–76. [Google Scholar] [CrossRef]
  13. Graham, C.N.; Watson, C.; Barlev, A.; Stevenson, M.; Dharnidharka, V.R. Mean lifetime survival estimates following solid organ transplantation in the US and UK. J. Med. Econ. 2022, 25, 230–237. [Google Scholar] [CrossRef] [PubMed]
  14. Samji, H.; Cescon, A.; Hogg, R.S.; Modur, S.P.; Althoff, K.N.; Buchacz, K.; Burchell, A.N.; Cohen, M.; Gebo, K.A.; Gill, M.J.; et al. Closing the gap: Increases in life expectancy among treated HIV-positive individuals in the United States and Canada. PLoS ONE 2013, 8, e81355. [Google Scholar] [CrossRef]
  15. Our New Approach to a Challenging Skin Cancer Statistic—The Skin Cancer Foundation. Available online: https://www.skincancer.org/blog/our-new-approach-to-a-challenging-skin-cancer-statistic/ (accessed on 13 May 2022).
  16. Cancer of Any Site—Cancer Stat Facts. Available online: https://seer.cancer.gov/statfacts/html/all.html (accessed on 14 July 2022).
  17. Goon, P.K.C.; Greenberg, D.C.; Igali, L.; Levell, N.J. Squamous Cell Carcinoma of the Skin has More than Doubled over the Last Decade in the UK. Acta Derm. Venereol. 2016, 96, 820–821. [Google Scholar] [CrossRef] [Green Version]
  18. Muzic, J.G.; Schmitt, A.R.; Wright, A.C.; Alniemi, D.T.; Zubair, A.S.; Olazagasti Lourido, J.M.; Sosa Seda, I.M.; Weaver, A.L.; Baum, C.L. Incidence and Trends of Basal Cell Carcinoma and Cutaneous Squamous Cell Carcinoma: A Population-Based Study in Olmsted County, Minnesota, 2000 to 2010. Mayo Clin. Proc. 2017, 92, 890–898. [Google Scholar] [CrossRef]
  19. Rogers, H.W.; Weinstock, M.A.; Feldman, S.R.; Coldiron, B.M. Incidence Estimate of Nonmelanoma Skin Cancer (Keratinocyte Carcinomas) in the U.S. Population, 2012. JAMA Dermatol. 2015, 151, 1081–1086. [Google Scholar] [CrossRef] [PubMed]
  20. Robsahm, T.E.; Helsing, P.; Veierød, M.B. Cutaneous squamous cell carcinoma in Norway 1963-2011: Increasing incidence and stable mortality. Cancer Med. 2015, 4, 472–480. [Google Scholar] [CrossRef]
  21. Lukowiak, T.M.; Aizman, L.; Perz, A.; Miller, C.J.; Sobanko, J.F.; Shin, T.M.; Giordano, C.N.; Higgins, H.W.; Etzkorn, J.R. Association of Age, Sex, Race, and Geographic Region with Variation of the Ratio of Basal Cell to Cutaneous Squamous Cell Carcinomas in the United States. JAMA Dermatol. 2020, 156, 1192–1198. [Google Scholar] [CrossRef]
  22. Mansouri, B.; Housewright, C.D. The Treatment of Actinic Keratoses-The Rule Rather Than the Exception. JAMA Dermatol. 2017, 153, 1200. [Google Scholar] [CrossRef]
  23. Melanoma of the Skin—Cancer Stat Facts. Available online: https://seer.cancer.gov/statfacts/html/melan.html (accessed on 15 July 2022).
  24. Zhang, W.; Zeng, W.; Jiang, A.; He, Z.; Shen, X.; Dong, X.; Feng, J.; Lu, H. Global, regional and national incidence, mortality and disability-adjusted life-years of skin cancers and trend analysis from 1990 to 2019: An analysis of the Global Burden of Disease Study 2019. Cancer Med. 2021, 10, 4905–4922. [Google Scholar] [CrossRef]
  25. National Comprehensive Cancer Network. Squamous Cell Skin Cancer (Version 5.2022). Available online: https://www.nccn.org/professionals/physician_gls/pdf/squamous.pdf (accessed on 13 May 2022).
  26. Chu, M.B.; Slutsky, J.B.; Dhandha, M.M.; Beal, B.T.; Armbrecht, E.S.; Walker, R.J.; Varvares, M.A.; Fosko, S.W. Evaluation of the definitions of “high-risk” cutaneous squamous cell carcinoma using the american joint committee on cancer staging criteria and national comprehensive cancer network guidelines. J. Skin Cancer 2014, 2014, 154340. [Google Scholar] [CrossRef] [PubMed]
  27. Rowe, D.E.; Carroll, R.J.; Day, C.L. Prognostic factors for local recurrence, metastasis, and survival rates in squamous cell carcinoma of the skin, ear, and lip. Implications for treatment modality selection. J. Am. Acad. Dermatol. 1992, 26, 976–990. [Google Scholar] [CrossRef]
  28. Farasat, S.; Yu, S.S.; Neel, V.A.; Nehal, K.S.; Lardaro, T.; Mihm, M.C.; Byrd, D.R.; Balch, C.M.; Califano, J.A.; Chuang, A.Y.; et al. A new American Joint Committee on Cancer staging system for cutaneous squamous cell carcinoma: Creation and rationale for inclusion of tumor (T) characteristics. J. Am. Acad. Dermatol. 2011, 64, 1051–1059. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Breuninger, H.; Brantsch, K.; Eigentler, T.; Häfner, H.-M. Comparison and evaluation of the current staging of cutaneous carcinomas. J. Dtsch. Dermatol. Ges. J. Ger. Soc. Dermatol. JDDG 2012, 10, 579–586. [Google Scholar] [CrossRef] [PubMed]
  30. 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] [PubMed] [Green Version]
  31. Amin, M.B.; Edge, S.B.; Greene, F.L.; Byrd, D.R.; Brookland, R.K.; Washington, M.K.; Gershenwald, J.E.; Compton, C.C.; Hess, K.R.; Sullivan, D.C.; et al. AJCC Cancer Staging Manual, 8th ed.; American Joint Committee on Cancer, American Cancer Society, CAPM-Managing editor, Laura, R.M., Eds.; Springer: Chicago, IL, USA, 2017; ISBN 978-3-319-40617-6. [Google Scholar]
  32. Schmitt, A.R.; Brewer, J.D.; Bordeaux, J.S.; Baum, C.L. Staging for cutaneous squamous cell carcinoma as a predictor of sentinel lymph node biopsy results: Meta-analysis of American Joint Committee on Cancer criteria and a proposed alternative system. JAMA Dermatol. 2014, 150, 19–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. 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]
  34. Venables, Z.C.; Tokez, S.; Hollestein, L.M.; Mooyaart, A.L.; van den Bos, R.R.; Rous, B.; Leigh, I.M.; Nijsten, T.; Wakkee, M. Validation of four cutaneous squamous cell carcinoma staging systems using nationwide data. Br. J. Dermatol. 2022, 186, 835–842. [Google Scholar] [CrossRef] [PubMed]
  35. Navarrete-Dechent, C.; Veness, M.J.; Droppelmann, N.; Uribe, P. High-risk cutaneous squamous cell carcinoma and the emerging role of sentinel lymph node biopsy: A literature review. J. Am. Acad. Dermatol. 2015, 73, 127–137. [Google Scholar] [CrossRef] [PubMed]
  36. Gore, S.M.; Shaw, D.; Martin, R.C.W.; Kelder, W.; Roth, K.; Uren, R.; Gao, K.; Davies, S.; Ashford, B.G.; Ngo, Q.; et al. Prospective study of sentinel node biopsy for high-risk cutaneous squamous cell carcinoma of the head and neck. Head Neck 2016, 38 (Suppl. S1), E884–E889. [Google Scholar] [CrossRef] [PubMed]
  37. Takahashi, A.; Imafuku, S.; Nakayama, J.; Nakaura, J.; Ito, K.; Shibayama, Y. Sentinel node biopsy for high-risk cutaneous squamous cell carcinoma. Eur. J. Surg. Oncol. J. Eur. Soc. Surg. Oncol. Br. Assoc. Surg. Oncol. 2014, 40, 1256–1262. [Google Scholar] [CrossRef]
  38. Wysong, A.; Newman, J.G.; Covington, K.R.; Kurley, S.J.; Ibrahim, S.F.; Farberg, A.S.; Bar, A.; Cleaver, N.J.; Somani, A.-K.; Panther, D.; et al. Validation of a 40-gene expression profile test to predict metastatic risk in localized high-risk cutaneous squamous cell carcinoma. J. Am. Acad. Dermatol. 2021, 84, 361–369. [Google Scholar] [CrossRef] [PubMed]
  39. Farberg, A.S.; Hall, M.A.; Douglas, L.; Covington, K.R.; Kurley, S.J.; Cook, R.W.; Dinehart, S.M. Integrating gene expression profiling into NCCN high-risk cutaneous squamous cell carcinoma management recommendations: Impact on patient management. Curr. Med. Res. Opin. 2020, 36, 1301–1307. [Google Scholar] [CrossRef] [PubMed]
  40. Work Group; Invited Reviewers; 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] [Green Version]
  41. Mulvaney, P.M.; Massey, P.R.; Yu, K.K.; Drinan, J.E.; Schmults, C.D. Differential Molecular Expression Patterns Associated with Metastasis in Cutaneous Squamous Cell Carcinoma: A Systematic Review and Meta-Analysis. J. Investig. Dermatol. 2021, 141, 2161–2169. [Google Scholar] [CrossRef] [PubMed]
  42. Cumsky, H.J.L.; Costello, C.M.; Zhang, N.; Butterfield, R.; Buras, M.R.; Schmidt, J.E.; Drenner, K.; Nelson, S.A.; Ochoa, S.A.; Baum, C.L.; et al. The prognostic value of inositol polyphosphate 5-phosphatase in cutaneous squamous cell carcinoma. J. Am. Acad. Dermatol. 2019, 80, 626–632.e1. [Google Scholar] [CrossRef]
  43. Chen, M.-K.; Cai, M.-Y.; Luo, R.-Z.; Tian, X.; Liao, Q.-M.; Zhang, X.-Y.; Han, J.-D. Overexpression of p300 correlates with poor prognosis in patients with cutaneous squamous cell carcinoma. Br. J. Dermatol. 2015, 172, 111–119. [Google Scholar] [CrossRef]
  44. Campos, M.A.; Macedo, S.; Fernandes, M.; Pestana, A.; Pardal, J.; Batista, R.; Vinagre, J.; Sanches, A.; Baptista, A.; Lopes, J.M.; et al. TERT promoter mutations are associated with poor prognosis in cutaneous squamous cell carcinoma. J. Am. Acad. Dermatol. 2019, 80, 660–669.e6. [Google Scholar] [CrossRef]
  45. Xu, R.; Cai, M.-Y.; Luo, R.-Z.; Tian, X.; Han, J.; Chen, M.-K. The Expression Status and Prognostic Value of Cancer Stem Cell Biomarker CD133 in Cutaneous Squamous Cell Carcinoma. JAMA Dermatol. 2016, 152, 305–311. [Google Scholar] [CrossRef] [Green Version]
  46. Piipponen, M.; Nissinen, L.; Kähäri, V.-M. Long non-coding RNAs in cutaneous biology and keratinocyte carcinomas. Cell. Mol. Life Sci. CMLS 2020, 77, 4601–4614. [Google Scholar] [CrossRef]
  47. Qiu, C.-G.; Shen, B.; Sun, X.-Q. Significant Biomarkers Identification Associated with Cutaneous Squamous Cell Carcinoma Progression. Int. J. Gen. Med. 2022, 15, 2347–2360. [Google Scholar] [CrossRef] [PubMed]
  48. Que, S.K.T.; Zwald, F.O.; Schmults, C.D. Cutaneous squamous cell carcinoma: Management of advanced and high-stage tumors. J. Am. Acad. Dermatol. 2018, 78, 249–261. [Google Scholar] [CrossRef]
  49. Fox, M.; Brown, M.; Golda, N.; Goldberg, D.; Miller, C.; Pugliano-Mauro, M.; Schmults, C.; Shin, T.; Stasko, T.; Xu, Y.G.; et al. Nodal staging of high-risk cutaneous squamous cell carcinoma. J. Am. Acad. Dermatol. 2019, 81, 548–557. [Google Scholar] [CrossRef] [PubMed]
  50. Baum, C.L.; Wright, A.C.; Martinez, J.-C.; Arpey, C.J.; Brewer, J.D.; Roenigk, R.K.; Otley, C.C. A new evidence-based risk stratification system for cutaneous squamous cell carcinoma into low, intermediate, and high risk groups with implications for management. J. Am. Acad. Dermatol. 2018, 78, 141–147. [Google Scholar] [CrossRef]
  51. MacFarlane, D.; Shah, K.; Wysong, A.; Wortsman, X.; Humphreys, T.R. The role of imaging in the management of patients with nonmelanoma skin cancer: Diagnostic modalities and applications. J. Am. Acad. Dermatol. 2017, 76, 579–588. [Google Scholar] [CrossRef]
  52. Tokez, S.; Koekelkoren, F.H.J.; Baatenburg de Jong, R.J.; Grünhagen, D.J.; Mooyaart, A.L.; Nijsten, T.; van der Lugt, A.; Wakkee, M. Assessment of the Diagnostic Accuracy of Baseline Clinical Examination and Ultrasonographic Imaging for the Detection of Lymph Node Metastasis in Patients with High-risk Cutaneous Squamous Cell Carcinoma of the Head and Neck. JAMA Dermatol. 2022, 158, 151–159. [Google Scholar] [CrossRef]
  53. Veness, M.J.; Morgan, G.J.; Palme, C.E.; Gebski, V. Surgery and adjuvant radiotherapy in patients with cutaneous head and neck squamous cell carcinoma metastatic to lymph nodes: Combined treatment should be considered best practice. Laryngoscope 2005, 115, 870–875. [Google Scholar] [CrossRef]
  54. Maher, J.M.; Schmults, C.D.; Murad, F.; Karia, P.S.; Benson, C.B.; Ruiz, E.S. Detection of subclinical disease with baseline and surveillance imaging in high-risk cutaneous squamous cell carcinomas. J. Am. Acad. Dermatol. 2020, 82, 920–926. [Google Scholar] [CrossRef]
  55. 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. Off. J. Am. Soc. Clin. Oncol. 2014, 32, 327–334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. 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] [Green Version]
  57. Bennardo, L.; Bennardo, F.; Giudice, A.; Passante, M.; Dastoli, S.; Morrone, P.; Provenzano, E.; Patruno, C.; Nisticò, S.P. Local Chemotherapy as an Adjuvant Treatment in Unresectable Squamous Cell Carcinoma: What Do We Know So Far? Curr. Oncol. 2021, 28, 2317–2325. [Google Scholar] [CrossRef]
  58. Pentangelo, G.; Nisticò, S.P.; Provenzano, E.; Cisale, G.Y.; Bennardo, L. Topical 5% Imiquimod Sequential to Surgery for HPV-Related Squamous Cell Carcinoma of the Lip. Medicina 2021, 57, 563. [Google Scholar] [CrossRef]
  59. 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] [Green Version]
  60. Jennings, L.; Schmults, C.D. Management of high-risk cutaneous squamous cell carcinoma. J. Clin. Aesthetic Dermatol. 2010, 3, 39–48. [Google Scholar]
  61. Barrett, T.L.; Greenway, H.T.; Massullo, V.; Carlson, C. Treatment of basal cell carcinoma and squamous cell carcinoma with perineural invasion. Adv. Dermatol. 1993, 8, 277–304, discussion 305. [Google Scholar]
  62. Calzavara-Pinton, P.G.; Venturini, M.; Sala, R.; Capezzera, R.; Parrinello, G.; Specchia, C.; Zane, C. Methylaminolaevulinate-based photodynamic therapy of Bowen’s disease and squamous cell carcinoma. Br. J. Dermatol. 2008, 159, 137–144. [Google Scholar] [CrossRef]
  63. Marmur, E.S.; Schmults, C.D.; Goldberg, D.J. A review of laser and photodynamic therapy for the treatment of nonmelanoma skin cancer. Dermatol. Surg. Off. Publ. Am. Soc. Dermatol. Surg. Al 2004, 30, 264–271. [Google Scholar] [CrossRef]
  64. Soleymani, T.; Abrouk, M.; Kelly, K.M. An Analysis of Laser Therapy for the Treatment of Nonmelanoma Skin Cancer. Dermatol. Surg. Off. Publ. Am. Soc. Dermatol. Surg. Al 2017, 43, 615–624. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Covadonga Martínez-González, M.; del Pozo, J.; Paradela, S.; Fernández-Jorge, B.; Fernández-Torres, R.; Fonseca, E. Bowen’s disease treated by carbon dioxide laser. A series of 44 patients. J. Dermatol. Treat. 2008, 19, 293–299. [Google Scholar] [CrossRef]
  66. Kim, H.-J.; Song, K.-H. Ablative fractional laser-assisted photodynamic therapy provides superior long-term efficacy compared with standard methyl aminolevulinate photodynamic therapy for lower extremity Bowen disease. J. Am. Acad. Dermatol. 2018, 79, 860–868. [Google Scholar] [CrossRef] [PubMed]
  67. Ko, D.Y.; Kim, K.H.; Song, K.H. A randomized trial comparing methyl aminolaevulinate photodynamic therapy with and without Er:YAG ablative fractional laser treatment in Asian patients with lower extremity Bowen disease: Results from a 12-month follow-up. Br. J. Dermatol. 2014, 170, 165–172. [Google Scholar] [CrossRef]
  68. Nguyen, B.T.; Gan, S.D.; Konnikov, N.; Liang, C.A. Treatment of superficial basal cell carcinoma and squamous cell carcinoma in situ on the trunk and extremities with ablative fractional laser-assisted delivery of topical fluorouracil. J. Am. Acad. Dermatol. 2015, 72, 558–560. [Google Scholar] [CrossRef] [PubMed]
  69. Wilmas, K.M.; Nguyen, Q.-B.; Patel, J.; Silapunt, S.; Migden, M.R. Treatment of advanced cutaneous squamous cell carcinoma: A Mohs surgery and dermatologic oncology perspective. Future Oncol. Lond. Engl. 2021, 17, 4971–4982. [Google Scholar] [CrossRef] [PubMed]
  70. Khansur, T.; Kennedy, A. Cisplatin and 5-fluorouracil for advanced locoregional and metastatic squamous cell carcinoma of the skin. Cancer 1991, 67, 2030–2032. [Google Scholar] [CrossRef]
  71. Shin, D.M.; Glisson, B.S.; Khuri, F.R.; Clifford, J.L.; Clayman, G.; Benner, S.E.; Forastiere, A.A.; Ginsberg, L.; Liu, D.; Lee, J.J.; et al. Phase II and biologic study of interferon alfa, retinoic acid, and cisplatin in advanced squamous skin cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2002, 20, 364–370. [Google Scholar] [CrossRef] [PubMed]
  72. DeConti, R.C. Chemotherapy of squamous cell carcinoma of the skin. Semin. Oncol. 2012, 39, 145–149. [Google Scholar] [CrossRef] [PubMed]
  73. Guthrie, T.H.; Porubsky, E.S.; Luxenberg, M.N.; Shah, K.J.; Wurtz, K.L.; Watson, P.R. Cisplatin-based chemotherapy in advanced basal and squamous cell carcinomas of the skin: Results in 28 patients including 13 patients receiving multimodality therapy. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 1990, 8, 342–346. [Google Scholar] [CrossRef] [PubMed]
  74. Aboul-Fettouh, N.; Morse, D.; Patel, J.; Migden, M.R. Immunotherapy and Systemic Treatment of Cutaneous Squamous Cell Carcinoma. Dermatol. Pract. Concept. 2021, 11, e2021169S. [Google Scholar] [CrossRef] [PubMed]
  75. Nottage, M.K.; Lin, C.; Hughes, B.G.M.; Kenny, L.; Smith, D.D.; Houston, K.; Francesconi, A. Prospective study of definitive chemoradiation in locally or regionally advanced squamous cell carcinoma of the skin. Head Neck 2017, 39, 679–683. [Google Scholar] [CrossRef]
  76. Sadek, H.; Azli, N.; Wendling, J.L.; Cvitkovic, E.; Rahal, M.; Mamelle, G.; Guillaume, J.C.; Armand, J.P.; Avril, M.F. Treatment of advanced squamous cell carcinoma of the skin with cisplatin, 5-fluorouracil, and bleomycin. Cancer 1990, 66, 1692–1696. [Google Scholar] [CrossRef]
  77. Edwards, L.; Berman, B.; Rapini, R.P.; Whiting, D.A.; Tyring, S.; Greenway, H.T.; Eyre, S.P.; Tanner, D.J.; Taylor, E.L.; Peets, E. Treatment of cutaneous squamous cell carcinomas by intralesional interferon alfa-2b therapy. Arch. Dermatol. 1992, 128, 1486–1489. [Google Scholar] [CrossRef] [PubMed]
  78. Lippman, S.M.; Parkinson, D.R.; Itri, L.M.; Weber, R.S.; Schantz, S.P.; Ota, D.M.; Schusterman, M.A.; Krakoff, I.H.; Gutterman, J.U.; Hong, W.K. 13-cis-retinoic acid and interferon alpha-2a: Effective combination therapy for advanced squamous cell carcinoma of the skin. J. Natl. Cancer Inst. 1992, 84, 235–241. [Google Scholar] [CrossRef] [PubMed]
  79. Kripke, M.L. Ultraviolet radiation and immunology: Something new under the sun—presidential address. Cancer Res. 1994, 54, 6102–6105. [Google Scholar] [PubMed]
  80. de Guillebon, E.; Roussille, P.; Frouin, E.; Tougeron, D. Anti program death-1/anti program death-ligand 1 in digestive cancers. World J. Gastrointest. Oncol. 2015, 7, 95–101. [Google Scholar] [CrossRef] [PubMed]
  81. Rizvi, N.A.; Hellmann, M.D.; Snyder, A.; Kvistborg, P.; Makarov, V.; Havel, J.J.; Lee, W.; Yuan, J.; Wong, P.; Ho, T.S.; et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015, 348, 124–128. [Google Scholar] [CrossRef] [Green Version]
  82. 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] [Green Version]
  83. Chan, T.A.; Wolchok, J.D.; Snyder, A. Genetic Basis for Clinical Response to CTLA-4 Blockade in Melanoma. N. Engl. J. Med. 2015, 373, 1984. [Google Scholar] [CrossRef] [Green Version]
  84. Lipson, E.J.; Forde, P.M.; Hammers, H.-J.; Emens, L.A.; Taube, J.M.; Topalian, S.L. Antagonists of PD-1 and PD-L1 in Cancer Treatment. Semin. Oncol. 2015, 42, 587–600. [Google Scholar] [CrossRef] [Green Version]
  85. Wherry, E.J. T cell exhaustion. Nat. Immunol. 2011, 12, 492–499. [Google Scholar] [CrossRef] [PubMed]
  86. 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] [Green Version]
  87. Clinicaltrials.gov. Regeneron Pharmaceuticals A First-in-Human Study of Repeat Dosing with REGN2810, a Monoclonal, Fully Human Antibody to Programmed Death-1 (PD-1), as Single Therapy and in Combination with Other Anti-Cancer Therapies in Patients with Advanced Malignancies; MD Anderson Cancer Center: Houston, TX, USA, 2020.
  88. Clinicaltrials.gov. Regeneron Pharmaceuticals A Phase 2 Study of REGN2810, a Fully Human Monoclonal Antibody to Programmed Death-1 (PD-1), in Patients with Advanced Cutaneous Squamous Cell Carcinoma; MD Anderson Cancer Center: Houston, TX, USA, 2021.
  89. FDA. Approves Cemiplimab-rwlc for Metastatic or Locally Advanced Cutaneous Squamous Cell Carcinoma|FDA. Available online: https://www.fda.gov/drugs/drug-approvals-and-databases/fda-approves-cemiplimab-rwlc-metastatic-or-locally-advanced-cutaneous-squamous-cell-carcinoma (accessed on 13 May 2022).
  90. 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] [PubMed]
  91. 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]
  92. WCM2021_Book_of_Abstracts. Available online: https://www.abstractserver.com/wcm2021/ebook/#320 (accessed on 13 May 2022).
  93. 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. Off. J. Am. Soc. Clin. Oncol. 2020, 38, 2916–2925. [Google Scholar] [CrossRef]
  94. Hughes, B.G.M.; Munoz-Couselo, E.; Mortier, L.; Bratland, Å.; Gutzmer, R.; Roshdy, O.; Mendoza, R.G.; Schachter, J.; Arance, A.; Grange, F.; et al. Phase 2 study of pembrolizumab (pembro) for locally advanced (LA) or recurrent/metastatic (R/M) cutaneous squamous cell carcinoma (cSCC): KEYNOTE-629 2021. In Proceedings of the 112th Annual Meeting of the American Association for Cancer Research, Orleans, LA, USA, 9–14 April 2021. [Google Scholar]
  95. Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [Green Version]
  96. Phan, G.Q.; Yang, J.C.; Sherry, R.M.; Hwu, P.; Topalian, S.L.; Schwartzentruber, D.J.; Restifo, N.P.; Haworth, L.R.; Seipp, C.A.; Freezer, L.J.; et al. Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma. Proc. Natl. Acad. Sci. USA 2003, 100, 8372–8377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Day, F.; Kumar, M.; Fenton, L.; Gedye, C. Durable Response of Metastatic Squamous Cell Carcinoma of the Skin to Ipilimumab Immunotherapy. J. Immunother. Hagerstown Md 1997 2017, 40, 36–38. [Google Scholar] [CrossRef]
  98. Sabbah, D.A.; Hajjo, R.; Sweidan, K. Review on Epidermal Growth Factor Receptor (EGFR) Structure, Signaling Pathways, Interactions, and Recent Updates of EGFR Inhibitors. Curr. Top. Med. Chem. 2020, 20, 815–834. [Google Scholar] [CrossRef] [PubMed]
  99. Galer, C.E.; Corey, C.L.; Wang, Z.; Younes, M.N.; Gomez-Rivera, F.; Jasser, S.A.; Ludwig, D.L.; El-Naggar, A.K.; Weber, R.S.; Myers, J.N. Dual inhibition of epidermal growth factor receptor and insulin-like growth factor receptor I: Reduction of angiogenesis and tumor growth in cutaneous squamous cell carcinoma. Head Neck 2011, 33, 189–198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. 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. Off. J. Am. Soc. Clin. Oncol. 2011, 29, 3419–3426. [Google Scholar] [CrossRef]
  101. Harari, P.M. Epidermal growth factor receptor inhibition strategies in oncology. Endocr. Relat. Cancer 2004, 11, 689–708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Foote, M.C.; McGrath, M.; Guminski, A.; Hughes, B.G.M.; Meakin, J.; Thomson, D.; Zarate, D.; Simpson, F.; Porceddu, S.V. Phase II study of single-agent panitumumab in patients with incurable cutaneous squamous cell carcinoma. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2014, 25, 2047–2052. [Google Scholar] [CrossRef]
  103. William, W.N.; 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]
  104. Gold, K.A.; Kies, M.S.; William, W.N.; 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] [PubMed]
  105. Fabbrocini, G.; Panariello, L.; Caro, G.; Cacciapuoti, S. Acneiform Rash Induced by EGFR Inhibitors: Review of the Literature and New Insights. Ski. Appendage Disord. 2015, 1, 31–37. [Google Scholar] [CrossRef] [Green Version]
  106. Wacker, B.; Nagrani, T.; Weinberg, J.; Witt, K.; Clark, G.; Cagnoni, P.J. Correlation between development of rash and efficacy in patients treated with the epidermal growth factor receptor tyrosine kinase inhibitor erlotinib in two large phase III studies. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2007, 13, 3913–3921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Hirsh, V. Managing treatment-related adverse events associated with egfr tyrosine kinase inhibitors in advanced non-small-cell lung cancer. Curr. Oncol. Tor. Ont. 2011, 18, 126–138. [Google Scholar] [CrossRef] [Green Version]
  108. Muñoz, N.M.; Williams, M.; Dixon, K.; Dupuis, C.; McWatters, A.; Avritscher, R.; Manrique, S.Z.; McHugh, K.; Murthy, R.; Tam, A.; et al. Influence of injection technique, drug formulation and tumor microenvironment on intratumoral immunotherapy delivery and efficacy. J. Immunother. Cancer 2021, 9, e001800. [Google Scholar] [CrossRef]
  109. Hannoodee, M.; Mittal, M. Methotrexate. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  110. Chan, E.S.L.; Cronstein, B.N. Methotrexate—How does it really work? Nat. Rev. Rheumatol. 2010, 6, 175–178. [Google Scholar] [CrossRef]
  111. Malaviya, A.N. Landmark papers on the discovery of methotrexate for the treatment of rheumatoid arthritis and other systemic inflammatory rheumatic diseases: A fascinating story. Int. J. Rheum. Dis. 2016, 19, 844–851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Bergón-Sendín, M.; Parra-Blanco, V.; Pulido-Pérez, A.; Nieto-Benito, L.M.; Rosell-Díaz, Á.M.; Suárez-Fernández, R. Histological findings after intralesional methotrexate treatment in cutaneous squamous cell carcinoma. Dermatol. Ther. 2020, 33, e14377. [Google Scholar] [CrossRef] [PubMed]
  113. Annest, N.M.; VanBeek, M.J.; Arpey, C.J.; Whitaker, D.C. Intralesional methotrexate treatment for keratoacanthoma tumors: A retrospective study and review of the literature. J. Am. Acad. Dermatol. 2007, 56, 989–993. [Google Scholar] [CrossRef]
  114. Bidaki, R.; Kian, M.; Owliaey, H.; Babaei Zarch, M.; Feysal, M. Accidental Chronic Poisoning with Methotrexate; Report of Two Cases. Emerg. Tehran Iran 2017, 5, e67. [Google Scholar]
  115. Longley, D.B.; Harkin, D.P.; Johnston, P.G. 5-fluorouracil: Mechanisms of action and clinical strategies. Nat. Rev. Cancer 2003, 3, 330–338. [Google Scholar] [CrossRef] [PubMed]
  116. Odom, R.B.; Goette, D.K. Treatment of keratoacanthomas with intralesional fluorouracil. Arch. Dermatol. 1978, 114, 1779–1783. [Google Scholar] [CrossRef]
  117. Good, L.M.; Miller, M.D.; High, W.A. Intralesional agents in the management of cutaneous malignancy: A review. J. Am. Acad. Dermatol. 2011, 64, 413–422. [Google Scholar] [CrossRef]
  118. Macdonald, J.S. Toxicity of 5-fluorouracil. Oncol. Williston Park N 1999, 13, 33–34. [Google Scholar]
  119. Newman, J.G.; Hall, M.A.; Kurley, S.J.; Cook, R.W.; Farberg, A.S.; Geiger, J.L.; Koyfman, S.A. Adjuvant therapy for high-risk cutaneous squamous cell carcinoma: 10-year review. Head Neck 2021, 43, 2822–2843. [Google Scholar] [CrossRef]
  120. Wang, J.T.; Palme, C.E.; Morgan, G.J.; Gebski, V.; Wang, A.Y.; Veness, M.J. Predictors of outcome in patients with metastatic cutaneous head and neck squamous cell carcinoma involving cervical lymph nodes: Improved survival with the addition of adjuvant radiotherapy. Head Neck 2012, 34, 1524–1528. [Google Scholar] [CrossRef]
  121. Trosman, S.J.; Zhu, A.; Nicolli, E.A.; Leibowitz, J.M.; Sargi, Z.B. High-Risk Cutaneous Squamous Cell Cancer of the Head and Neck: Risk Factors for Recurrence and Impact of Adjuvant Treatment. Laryngoscope 2021, 131, E136–E143. [Google Scholar] [CrossRef]
  122. Oun, R.; Moussa, Y.E.; Wheate, N.J. The side effects of platinum-based chemotherapy drugs: A review for chemists. Dalton Trans. Camb. Eng. 2003 2018, 47, 6645–6653. [Google Scholar] [CrossRef]
  123. Harwood, C.A.; Leedham-Green, M.; Leigh, I.M.; Proby, C.M. Low-dose retinoids in the prevention of cutaneous squamous cell carcinomas in organ transplant recipients: A 16-year retrospective study. Arch. Dermatol. 2005, 141, 456–464. [Google Scholar] [CrossRef] [Green Version]
  124. George, R.; Weightman, W.; Russ, G.R.; Bannister, K.M.; Mathew, T.H. Acitretin for chemoprevention of non-melanoma skin cancers in renal transplant recipients. Australas. J. Dermatol. 2002, 43, 269–273. [Google Scholar] [CrossRef]
  125. McKenna, D.B.; Murphy, G.M. Skin cancer chemoprophylaxis in renal transplant recipients: 5 years of experience using low-dose acitretin. Br. J. Dermatol. 1999, 140, 656–660. [Google Scholar] [CrossRef]
  126. Gibson, G.E.; O’Grady, A.; Kay, E.W.; Leader, M.; Murphy, G.M. Langerhans cells in benign, premalignant and malignant skin lesions of renal transplant recipients and the effect of retinoid therapy. J. Eur. Acad. Dermatol. Venereol. 1998, 10, 130–136. [Google Scholar] [CrossRef]
  127. Bavinck, J.N.; Tieben, L.M.; Van der Woude, F.J.; Tegzess, A.M.; Hermans, J.; ter Schegget, J.; Vermeer, B.J. Prevention of skin cancer and reduction of keratotic skin lesions during acitretin therapy in renal transplant recipients: A double-blind, placebo-controlled study. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 1995, 13, 1933–1938. [Google Scholar] [CrossRef]
  128. Shuttleworth, D.; Marks, R.; Griffin, P.J.; Salaman, J.R. Treatment of cutaneous neoplasia with etretinate in renal transplant recipients. Q. J. Med. 1988, 68, 717–725. [Google Scholar]
  129. Nijsten, T.E.C.; Stern, R.S. Oral retinoid use reduces cutaneous squamous cell carcinoma risk in patients with psoriasis treated with psoralen-UVA: A nested cohort study. J. Am. Acad. Dermatol. 2003, 49, 644–650. [Google Scholar] [CrossRef]
  130. Weinstock, M.A.; Bingham, S.F.; Digiovanna, J.J.; Rizzo, A.E.; Marcolivio, K.; Hall, R.; Eilers, D.; Naylor, M.; Kirsner, R.; Kalivas, J.; et al. Tretinoin and the prevention of keratinocyte carcinoma (Basal and squamous cell carcinoma of the skin): A veterans affairs randomized chemoprevention trial. J. Investig. Dermatol. 2012, 132, 1583–1590. [Google Scholar] [CrossRef] [Green Version]
  131. Tangrea, J.A.; Edwards, B.K.; Taylor, P.R.; Hartman, A.M.; Peck, G.L.; Salasche, S.J.; Menon, P.A.; Benson, P.M.; Mellette, J.R.; Guill, M.A. Long-term therapy with low-dose isotretinoin for prevention of basal cell carcinoma: A multicenter clinical trial. Isotretinoin-Basal Cell Carcinoma Study Group. J. Natl. Cancer Inst. 1992, 84, 328–332. [Google Scholar] [CrossRef]
  132. Levine, N.; Moon, T.E.; Cartmel, B.; Bangert, J.L.; Rodney, S.; Dong, Q.; Peng, Y.M.; Alberts, D.S. Trial of retinol and isotretinoin in skin cancer prevention: A randomized, double-blind, controlled trial. Southwest Skin Cancer Prevention Study Group. Cancer Epidemiol. Biomark. Prev. Publ. Am. Assoc. Cancer Res. Cosponsored Am. Soc. Prev. Oncol. 1997, 6, 957–961. [Google Scholar]
  133. Moon, T.E.; Levine, N.; Cartmel, B.; Bangert, J.L.; Rodney, S.; Dong, Q.; Peng, Y.M.; Alberts, D.S. Effect of retinol in preventing squamous cell skin cancer in moderate-risk subjects: A randomized, double-blind, controlled trial. Southwest Skin Cancer Prevention Study Group. Cancer Epidemiol. Biomark. Prev. Publ. Am. Assoc. Cancer Res. Cosponsored Am. Soc. Prev. Oncol. 1997, 6, 949–956. [Google Scholar]
  134. Weinstock, M.A.; Thwin, S.S.; Siegel, J.A.; Marcolivio, K.; Means, A.D.; Leader, N.F.; Shaw, F.M.; Hogan, D.; Eilers, D.; Swetter, S.M.; et al. Chemoprevention of Basal and Squamous Cell Carcinoma with a Single Course of Fluorouracil, 5%, Cream: A Randomized Clinical Trial. JAMA Dermatol. 2018, 154, 167–174. [Google Scholar] [CrossRef] [Green Version]
  135. Chen, A.C.; Martin, A.J.; Choy, B.; Fernández-Peñas, P.; Dalziell, R.A.; McKenzie, C.A.; Scolyer, R.A.; Dhillon, H.M.; Vardy, J.L.; Kricker, A.; et al. A Phase 3 Randomized Trial of Nicotinamide for Skin-Cancer Chemoprevention. N. Engl. J. Med. 2015, 373, 1618–1626. [Google Scholar] [CrossRef] [Green Version]
  136. Mainville, L.; Smilga, A.-S.; Fortin, P.R. Effect of Nicotinamide in Skin Cancer and Actinic Keratoses Chemoprophylaxis, and Adverse Effects Related to Nicotinamide: A Systematic Review and Meta-Analysis. J. Cutan. Med. Surg. 2022, 26, 297–308. [Google Scholar] [CrossRef] [PubMed]
  137. Lewis, K.G.; Lewis, M.D.; Robinson-Bostom, L.; Pan, T.D. Inflammation of actinic keratoses during capecitabine therapy. Arch. Dermatol. 2004, 140, 367–368. [Google Scholar] [CrossRef]
  138. Schauder, D.M.; Kim, J.; Nijhawan, R.I. Evaluation of the Use of Capecitabine for the Treatment and Prevention of Actinic Keratoses, Squamous Cell Carcinoma, and Basal Cell Carcinoma: A Systematic Review. JAMA Dermatol. 2020, 156, 1117–1124. [Google Scholar] [CrossRef]
  139. Jirakulaporn, T.; Endrizzi, B.; Lindgren, B.; Mathew, J.; Lee, P.K.; Dudek, A.Z. Capecitabine for skin cancer prevention in solid organ transplant recipients. Clin. Transplant. 2011, 25, 541–548. [Google Scholar] [CrossRef]
  140. Zhang, B.; Liang, X.; Ye, L.; Wang, Y. No chemopreventive effect of nonsteroidal anti-inflammatory drugs on nonmelanoma skin cancer: Evidence from meta-analysis. PLoS ONE 2014, 9, e96887. [Google Scholar] [CrossRef] [PubMed]
  141. Willey, A.; Mehta, S.; Lee, P.K. Reduction in the incidence of squamous cell carcinoma in solid organ transplant recipients treated with cyclic photodynamic therapy. Dermatol. Surg. Off. Publ. Am. Soc. Dermatol. Surg. Al 2010, 36, 652–658. [Google Scholar] [CrossRef]
  142. Innovent Biologics (Suzhou) Co., Ltd. A Phase Ib/II, Open-Label, Single Arm Study to Evaluate the Safety and Efficacy of IBI318 in Participants with Advanced Cutaneous Squamous Cell Carcinoma; clinicaltrials.gov: Bethesda, MD, USA, 2020.
  143. Shattuck Labs, Inc. Phase 1 Dose Escalation and Dose Expansion Study of an Agonist Redirected Checkpoint Fusion Protein, SL-279252 (PD1-Fc-OX40L), in Subjects with Advanced Solid Tumors or Lymphomas; clinicaltrials.gov: Bethesda, MD, USA, 2022.
  144. Checkpoint Therapeutics, Inc. A Phase 1, Open-Label, Multicenter, Dose-Escalation Study of CK-301 Administered Intravenously as a Single Agent to Subjects with Advanced Cancers; clinicaltrials.gov: Bethesda, MD, USA, 2022.
  145. SOTIO Biotech AG. A Phase 2, Open-label, Single-arm, Multicenter Study of SOT101 in Combination with Pembrolizumab to Evaluate the Efficacy and Safety in Patients with Selected Advanced/Refractory Solid Tumors; clinicaltrials.gov: Bethesda, MD, USA, 2022.
  146. InflaRx GmbH. Open Label, Multicenter Phase II Study of the C5a Antibody IFX-1 Alone or IFX-1 + Pembrolizumab in Patients with PD-1 or PD-L1 Resistant/Refractory Locally Advanced or Metastatic Cutaneous Squamous Cell Carcinoma (cSCC); clinicaltrials.gov: Bethesda, MD, USA, 2021.
  147. CureVac AG. Phase I Study of Intratumoral CV8102 in Patients with Advanced Melanoma, Squamous Cell Carcinoma of the Skin, Squamous Cell Carcinoma of the Head and Neck, or Adenoid Cystic Carcinoma; clinicaltrials.gov: Bethesda, MD, USA, 2021.
  148. NeoImmuneTech. A Phase 1b/2a, Open Label Study to Evaluate Anti-tumor Efficacy and Safety of rhIL-7-hyFc (NT-I7) in Combination with Anti-PD-L1 (Atezolizumab) in Patients with Anti-PD-1/PD-L1 naïve or Relapsed/Refractory High-risk Skin Cancers; clinicaltrials.gov: Bethesda, MD, USA, 2021.
  149. Sanofi, A. Phase 1/2 Non-Randomized, Open-Label, Multi-Cohort, Multi-Center Study Assessing the Clinical Benefit of SAR444245 (THOR-707) Combined with Cemiplimab for the Treatment of Participants with Advanced Unresectable or Metastatic Skin Cancers; clinicaltrials.gov: Bethesda, MD, USA, 2022.
  150. Checkmate Pharmaceuticals. A Multicenter, Open-label, Phase 2 Study of Intratumoral CMP-001 in Combination with an Intravenous PD-1-Blocking Antibody in Subjects with Selected Types of Advanced or Metastatic Cancer; clinicaltrials.gov: Bethesda, MD, USA, 2022.
  151. Bicara Therapeutics. First-in-Human, Phase 1/1b, Open-label, Multicenter Study of Bifunctional EGFR/TGFβ Fusion Protein BCA101 Monotherapy and in Combination Therapy in Patients with EGFR-Driven Advanced Solid Tumors; clinicaltrials.gov: Bethesda, MD, USA, 2022.
  152. Nektar Therapeutics. A Phase 1b/2, Open-label, Multicenter, Dose Escalation and Dose Expansion Study of NKTR-255 Monotherapy or in Combination with Cetuximab as a Salvage Regimen for Solid Tumors; clinicaltrials.gov: Bethesda, MD, USA, 2022.
  153. Regeneron Pharmaceuticals. A Phase 1 Study of Pre-Operative Cemiplimab (REGN2810), Administered Intralesionally, for Patients with Recurrent Cutaneous Squamous Cell Carcinoma (CSCC); clinicaltrials.gov: Bethesda, MD, USA, 2022.
  154. Codiak BioSciences. A First-in-Human Study of CDK-002 (exoSTING) in Subjects with Advanced/Metastatic, Recurrent, Injectable Solid Tumors, with Emphasis on Squamous Cell Carcinoma of the Head and Neck, Triple Negative Breast Cancer, Anaplastic Thyroid Carcinoma, and Cutaneous Squamous Cell Carcinoma; clinicaltrials.gov: Bethesda, MD, USA, 2022.
  155. Sirnaomics. An Open Label, Dose Escalation Study to Evaluate the Safety and Efficacy of Intralesional Injection of STP705 in Adult Patients with Cutaneous Squamous Cell Carcinoma in Situ (isSCC); clinicaltrials.gov: Bethesda, MD, USA, 2022.
  156. Shattuck Labs, Inc. Phase 1 Dose Escalation Study of the Agonist Redirected Checkpoint, SL-172154 (SIRPα-Fc-CD40L), Administered Intratumorally in Subjects with Cutaneous Squamous Cell Carcinoma or Squamous Cell Carcinoma of the Head and Neck; clinicaltrials.gov: Bethesda, MD, USA, 2022.
  157. Rakuten Medical, Inc. An Open-Label Study Using ASP-1929 Photoimmunotherapy in Combination with Anti-PD1 Therapy in EGFR Expressing Advanced Solid Tumors; clinicaltrials.gov: Bethesda, MD, USA, 2022.
  158. Rakuten Medical, Inc. A Phase 1 First-in-Human, Drug-dose Escalation Study of RM-1995 Photoimmunotherapy, as Monotherapy or Combined with Pembrolizumab, in Patients with Advanced Cutaneous Squamous Cell Carcinoma or with Head and Neck Squamous Cell Carcinoma; clinicaltrials.gov: Bethesda, MD, USA, 2022.
  159. PhD, O.E.A. Phase Ib/II Study of Micro-Needle Array Containing Doxorubicin in Immune Competent or Immune-suppressed Patients with Cutaneous Squamous Cell Carcinoma; clinicaltrials.gov: Bethesda, MD, USA, 2022.
  160. Alpha Tau Medical Ltd. A Prospective International Multicenter, Pivotal, Single Arm, Open Label Clinical Study to Assess the Efficacy and Safety of Intratumoral Alpha DaRT224 for the Treatment of Patients with Recurrent Cutaneous Squamous Cell Carcinoma; clinicaltrials.gov: Bethesda, MD, USA, 2022.
  161. Morphogenesis, Inc. Phase 1 Trial of IFx-Hu2.0 to Evaluate Safety in Patients with Skin Cancer; clinicaltrials.gov: Bethesda, MD, USA, 2022.
  162. Morphogenesis, Inc. Phase 1 Trial of Intralesional Immunotherapy with IFx-Hu2.0 Vaccine in Patients with Advanced Non-Melanoma Skin Cancers; clinicaltrials.gov: Bethesda, MD, USA, 2022.
  163. University of Arizona. A Single Arm Phase 2 Study of Talimogene Laherparepvec in Patients with Cutaneous Squamous Cell Cancer; clinicaltrials.gov: Bethesda, MD, USA, 2022.
  164. National Cancer Institute (NCI). A Phase II Study of Talimogene Laherparepvec Followed by Talimogene Laherparepvec + Nivolumab in Refractory T Cell and NK Cell Lymphomas, Cutaneous Squamous Cell Carcinoma, Merkel Cell Carcinoma, and Other Rare Skin Tumors; clinicaltrials.gov: Bethesda, MD, USA, 2022.
  165. MD, A.B. A Phase 1 Study of Talimogene Laherparepvec and Panitumumab in Patients with Locally Advanced Squamous Cell Carcinoma of the Skin (SCCS); clinicaltrials.gov: Bethesda, MD, USA, 2022.
  166. Replimune Inc. A Randomized, Controlled, Open-Label, Phase 2 Study of Cemiplimab as a Single Agent and in Combination with RP1 in Patients with Advanced Cutaneous Squamous Cell Carcinoma; clinicaltrials.gov: Bethesda, MD, USA, 2022.
  167. Turnstone Biologics, Corp. A Phase 1/2a, Multicenter, Open-label Trial of TBio-6517, an Oncolytic Vaccinia Virus, Administered Alone and in Combination with Pembrolizumab, in Patients with Advanced Solid Tumors; clinicaltrials.gov: Bethesda, MD, USA, 2022.
  168. Murakami, N.; Mulvaney, P.; Danesh, M.; Abudayyeh, A.; Diab, A.; Abdel-Wahab, N.; Abdelrahim, M.; Khairallah, P.; Shirazian, S.; Kukla, A.; et al. A multi-center study on safety and efficacy of immune checkpoint inhibitors in cancer patients with kidney transplant. Kidney Int. 2021, 100, 196–205. [Google Scholar] [CrossRef] [PubMed]
  169. Hanna, G.J. Safety and Efficacy of Cemiplimab (PD-1 Blockade) in Selected Organ Transplant Recipients with Advanced Cutaneous Squamous Cell Carcinoma (CONTRAC); clinicaltrials.gov: Bethesda, MD, USA, 2021.
  170. Replimune Inc. An Open-Label, Multicenter, Phase 1B/2 Study of RP1 in Solid Organ Transplant Recipients with Advanced Cutaneous Malignancies; clinicaltrials.gov: Bethesda, MD, USA, 2022.
  171. ClinicalTrials.gov. Immunotherapy Multi-Omics Specimen Protocol A; clinicaltrials.gov: Bethesda, MD, USA, 2021.
  172. Kyte, J.A. MITRIC: Microbiota Transplant to Cancer Patients Who Have Failed Immunotherapy Using Faeces from Clincal Responders; clinicaltrials.gov: Bethesda, MD, USA, 2022.
  173. Regeneron Pharmaceuticals. Cemiplimab Survivorship Epidemiology (CASE) Study; clinicaltrials.gov: Bethesda, MD, USA, 2022.
  174. University of Erlangen-Nürnberg Medical School. Clinical Benefit and Biomarker Analysis of Combination of PD-1/PD-L1 Immune Checkpoint Inhibitors and Radiotherapy in NSCLC, HNSCC and Other Solid Tumors; clinicaltrials.gov: Bethesda, MD, USA, 2021.
Table 1. National Comprehensive Cancer Network features of high-risk and very high-risk cutaneous squamous cell carcinoma (May 2022); PNI, perineural invasion.
Table 1. National Comprehensive Cancer Network features of high-risk and very high-risk cutaneous squamous cell carcinoma (May 2022); PNI, perineural invasion.
High-Risk cSCCVery High-Risk cSCC
Tumors on the head, neck, hands, feet, pretibial, and anogenital region or trunk and extremities with diameter ≥ 2 cmTumors with diameter ≥ 4 cm on any location
Acantholytic, adenosquamous, or metaplastic subtypes, with PNIPoorly differentiated, desmoplastic, >6 mm thickness, or invasion beyond fat
PNI ≥ 0.1 mmPNI with tumor within nerve sheath of a nerve lying deeper than the dermis or ≥0.1 mm
Recurrent tumorLymphatic or vascular involvement
History of immunosuppression, prior site of radiotherapy, rapidly growing tumor, neurologic symptoms
Table 2. Summary of four staging systems for cutaneous squamous cell carcinoma. PNI, perineural invasion.
Table 2. Summary of four staging systems for cutaneous squamous cell carcinoma. PNI, perineural invasion.
Staging SystemStageRisk FactorsHigh-Risk Factors
(If Applicable)
AJCC7T1Tumor diameter ≤ 2 cm with <2 high-risk factors>2 mm thickness
Clark level ≥ 4
PNI
Primary site ear or lip
Poorly differentiated
T2Tumor diameter > 2 cm, or any size with ≥2 high-risk factors
T3Tumor with invasion of orbit, maxilla, mandible, or temporal bone
T4Tumor with invasion of other bone, or direct PNI of skull base
AJCC8 T1Tumor diameter < 2 cm
T2Tumor diameter ≥ 2 cm and <4 cm
T3Tumor diameter ≥ 4 cm, or minor bone erosion, or PNI, or deep invasion
T4Tumor with gross cortical bone/marrow invasion
BWHT1No high-risk factorsTumor diameter ≥ 2 cm
PNI ≥ 0.1 mm
Poorly differentiated
Tumor invasion beyond fat
T2a1 high-risk factor
T2b2–3 high-risk factors
T3≥4 high-risk factors, or bone invasion
Brueninger et al.Clinical stage (cT)Low risk: Tumor diameter ≤ 2 cm
High risk: Tumor diameter > 2 cm
Pathological stage (pT)No risk: Tumor thickness ≤ 2 mm
Low risk: Tumor thickness > 2 mm and ≤6 mm
High risk: Tumor thickness > 6 mm
Co-risk factorsImmunosuppression
Desmoplastic type or poor differentiation
Primary site ear
Table 3. Recurrence of localized cutaneous squamous cell carcinoma after surgery; cSCC, cutaneous squamous cell carcinoma.
Table 3. Recurrence of localized cutaneous squamous cell carcinoma after surgery; cSCC, cutaneous squamous cell carcinoma.
Primary cSCCRecurrent cSCC
Mohs3.1%10.9%
Standard Excision10.0%23.3%
Table 4. Expanded follow-up of phase 2 cemiplimab data; ORR, objective response rate; PFS, progression free survival (in months).
Table 4. Expanded follow-up of phase 2 cemiplimab data; ORR, objective response rate; PFS, progression free survival (in months).
GroupCohortDosingPatients (n)ORRMedian PFS
1Metastatic cSCC3 mg/kg IV q2 weeks5950.8%18.4
2Locally advanced cSCC3 mg/kg IV q2 weeks7844.9%18.5
3Metastatic cSCC3 mg/kg IV q3 weeks5646.4%21.7
Total 19347.2%18.5
Table 5. Expanded follow-up of phase 2 pembrolizumab data; ORR, objective response rate; PFS, progression free survival (in months); NR, not reached.
Table 5. Expanded follow-up of phase 2 pembrolizumab data; ORR, objective response rate; PFS, progression free survival (in months); NR, not reached.
CohortDosingPatients (n)ORRMedian PFS
Locally advanced cSCC200 mg IV q3 weeks5450.0%NR
Recurrent/metastatic cSCC200 mg IV q3 weeks10535.2%5.7
Total 15940.3%NR
Table 6. Prospective study summaries for targeted therapy of cutaneous squamous cell carcinoma; ORR, objective response rate; CR, complete response; PR, partial response; PFS, progression free survival (in months); OS, overall survival (in months). * All patients treated as second line.
Table 6. Prospective study summaries for targeted therapy of cutaneous squamous cell carcinoma; ORR, objective response rate; CR, complete response; PR, partial response; PFS, progression free survival (in months); OS, overall survival (in months). * All patients treated as second line.
RouteTherapyORRCRPRMedian PFSMedian OS
OralErlotinib10%0%10%4.713
Gefitinib16%0%16%3.812.9
Intravenous Cetuximab28%6%22%4.18.1
Panitumumab31% *13% *19% *8 *11 *
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Burns, C.; Kubicki, S.; Nguyen, Q.-B.; Aboul-Fettouh, N.; Wilmas, K.M.; Chen, O.M.; Doan, H.Q.; Silapunt, S.; Migden, M.R. Advances in Cutaneous Squamous Cell Carcinoma Management. Cancers 2022, 14, 3653. https://doi.org/10.3390/cancers14153653

AMA Style

Burns C, Kubicki S, Nguyen Q-B, Aboul-Fettouh N, Wilmas KM, Chen OM, Doan HQ, Silapunt S, Migden MR. Advances in Cutaneous Squamous Cell Carcinoma Management. Cancers. 2022; 14(15):3653. https://doi.org/10.3390/cancers14153653

Chicago/Turabian Style

Burns, Carrick, Shelby Kubicki, Quoc-Bao Nguyen, Nader Aboul-Fettouh, Kelly M. Wilmas, Olivia M. Chen, Hung Quoc Doan, Sirunya Silapunt, and Michael R. Migden. 2022. "Advances in Cutaneous Squamous Cell Carcinoma Management" Cancers 14, no. 15: 3653. https://doi.org/10.3390/cancers14153653

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