[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
Next Article in Journal
Clinical, Dermoscopic, and Molecular Features of Acantholytic Squamous Cell Carcinoma: A Systematic Review
Previous Article in Journal
Neutrophil-to-Lymphocyte and Platelet-to-Lymphocyte Ratio: Side by Side with Molecular Mutations in Patients with Non-Small Cell Lung Cancer—The INOLUNG Study
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Mutational and Microenvironmental Landscape of Cutaneous Squamous Cell Carcinoma: A Review

1
Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
2
Division of Hematology-Oncology, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
3
Department of Otolaryngology-Head & Neck Surgery, University of California San Diego, La Jolla, CA 92093, USA
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(16), 2904; https://doi.org/10.3390/cancers16162904
Submission received: 16 July 2024 / Revised: 9 August 2024 / Accepted: 19 August 2024 / Published: 21 August 2024
(This article belongs to the Section Tumor Microenvironment)

Abstract

:

Simple Summary

Cutaneous squamous cell carcinoma (cSCC) is a complex disease arising from an interplay of UV-induced DNA damage, genetic mutations, and alterations in the tumor microenvironment. While most cSCC cases can be treated with surgery, metastatic and locally advanced forms have a significantly worse prognosis and limited treatment options. The high mutational burden in cSCC makes it challenging to identify true driver alterations. Beyond genetics, the tumor microenvironment and its interaction with the immune system play a critical role in cSCC progression, evidenced by differences observed between immunocompromised and immunocompetent patients. In recent years, new therapeutic approaches have emerged, including immunotherapy and EGFR inhibitors, which have shown efficacy in treating advanced cSCC. Understanding the underlying mechanisms driving cSCC is crucial for developing novel and more effective therapies. This review aims to provide a comprehensive overview of the mutational landscape and microenvironmental factors in cSCC.

Abstract

Cutaneous squamous cell carcinoma (cSCC) manifests through the complex interactions of UV-induced DNA damage, genetic mutations, and alterations in the tumor microenvironment. A high mutational burden is present in cSCC, as well as both cSCC precursors and normal skin, making driver genes difficult to differentiate. Despite this, several key driver genes have been identified, including TP53, the NOTCH family, CDKN2A, PIK3CA, and EGFR. In addition to mutations, the tumor microenvironment and the manipulation and evasion of the immune system play a critical role in cSCC progression. Novel therapeutic approaches, such as immunotherapy and EGFR inhibitors, have been used to target these dysregulations, and have shown promise in treating advanced cSCC cases, emphasizing the need for targeted interventions considering both genetic and microenvironmental factors for improved patient outcomes.

1. Introduction

Cutaneous squamous cell carcinoma (cSCC) is the second most common subtype of non-melanoma skin cancer, as it is responsible for 20% of all skin cancers [1,2]. Major risk factors include ultraviolet (UV) radiation exposure, male sex, pale complexion, xeroderma pigmentosum, and immunosuppression [2,3,4,5,6,7]. Treatment depends on the level of aggressiveness and metastatic status, with non-metastatic cases usually successfully removed via surgical excision [6]. Metastatic cSCC, which accounts for only 5% of cases, is associated with a worse prognosis and often requires a combinatory treatment of surgery, radiation, and systemic therapy [2,6,8].
The current understanding of cSCC development is multifaceted, with the tumorigenesis associated with it contributing to a disruption of epidermal homeostasis, stemming from alterations at various points of the development process; these alterations include UV-induced DNA damage, gene mutations, and tumor microenvironment changes [9,10]. The recent success in the treatment of cSCC with immune checkpoint inhibitors also demonstrates the important interplay between cSCC and the tumor immune microenvironment [11,12].
This review presents an overview of the literature on cSCC from a genetic and tissue microenvironmental perspective to improve our current understanding of this disease and highlight potential novel therapeutic targets for future study.

2. Genetic Landscape

2.1. UV-Signature Mutations

Ultraviolet exposure, specifically UVB exposure, is the most important, preventable risk factor in the development of cSCC, as it induces DNA mutations within the skin [8,13]. Normally, these mutations are repaired via a process known as nucleotide excision repair (NER), but when NER process is defective, cSCC development can occur [13]. The most frequent UV-signature mutations are C > T substitution mutations at dipyrimidine sites, often accompanied by CC > TT dinucleotide substitutions on the non-transcribed strand of many genes [1,14,15]. C > A (G > T) is another described mutational signature seen in cSCC with low mutational burden [1,15]. Recent research has suggested a novel UV mutational signature, revealing that T > C substitutions are also a frequent result of UVB exposure, with 5’-ATT-3’ and 5′-ATG-3′ as reported sequence motifs [16].

2.2. Mutations in Normal Skin

Identifying UV signatures alone is insufficient in understanding the trajectory from cSCC to tumorigenesis. Interestingly, many of mutations in cSCC are also seen in normal sun-exposed skin [15]. Genes from the NOTCH family (NOTCH1, NOTCH2, and NOTCH3) are frequently mutated in normal skin, with alterations in NOTCH genes having an average of 83 driver mutations per square centimeter of normal skin, resulting in significant protein alterations [15]. TP53 is also frequently mutated in normal skin, with a rate of 9.5 driver mutations per square centimeter of skin [15]. Furthermore, mutations in FAT1 and well-known oncogenes like KRAS, NRAS, and HRAS have also been identified in normal skin [15].
This high mutation occurrence in normal skin makes differentiating somatic driver mutations from passenger mutations quite difficult [1,5]. Other limitations include tumor heterogeneity, neoplastic cell content, mean sequencing coverage, and variable sequencing coverage across tumors [5]. Fortunately, these limitations can be addressed by using large cohort sizes [1], conducting meta-analysis [5], having strict quality control measures, and understanding the mutational burdens in normal skin [5,15].

2.3. Hereditary Mutations

While most cSCCs arise from somatic mutations, certain hereditary diseases can also contribute to the development of cSCC. Xeroderma pigmentosum (XP) is a genetic condition known to predispose patients to cSCC, basal cell carcinoma (BCC), and melanoma [3,17,18]. XP is an autosomal-recessive disorder that causes extreme sensitivity to UV exposure, due to mutations in the NER pathway, which hinder the DNA’s ability to repair itself after UV damage [5,18]. As a result, XP patients have an extremely high risk for developing skin cancer, with patients under 20 having a 10,000-fold increased risk of developing cSCCs and BCCs and a 2,000-fold increase for melanoma [3].

2.4. Mutations in Pre-Cancer

The primary cSCC precursor is actinic keratosis (AK), an in situ neoplasm that originates from mutated clonal DNA in keratinocytes [19,20,21]. AKs manifest as macules, papules, or hyperkeratotic plaques that are identified via visual and palpation inspections [20]. Both AK and cSCC share the same risk factors, the most important being UV exposure, as well as male sex, fair skin, and old age [19,20]. However, only some AK cases progress into cSCC, and the exact pathway to malignancy is not well understood [20,21].
The ability to define clear genetic criteria for the malignant transformation of AK to cSCC is limited by the high-UV mutational burden [20,21]. Both cSCC and AK demonstrate many genetic similarities, including mutations within driver genes and mutational signatures [21]. TP53 is renowned for being an early mutational event in both AK and cSCC, usually as nonsense or missense mutations with specific hotspots R248W and G245D found in normal sun-exposed skin [1,5,8,19,20,21,22,23,24]. Other genetic alterations in genes NOTCH1, NOTCH2, FAT1, and KMT2C and mutational signatures C > T and CC > TT have been found in both AK and cSCC; however, whether or not these mutated genes and UV signatures can reliably predict the trajectory from AK to cSCC progression is still being investigated [21]. One study found that cSCC is most associated with common and hypertrophic AK types, with low p53 immunopositivity predicting if common-type AK will develop into cSCC [19]. While 65% of SCCs are derived from AKs, only about 0.6–2.57% of AK cases progress into cSCC, partly because AK can regress [25,26].
Although, AK and cSCC are very similar in their mutational burden values, 43.5 mutations per megabase DNA and 50 mutations per megabase DNA respectively, cSCC experiences more intra-sample heterogeneity than AK [21,27]. They also differ in terms of the many pathways they involve, which are activated in cSCC but not activated in AK, such as the PI3K/AKT/mTOR and RAS/RAF/MEK/ERK pathways [28,29,30,31,32]. Additionally, immune system signaling pathways like those for TGFβ, adipocytokine, GnRH, and insulin signaling are more highly mutated in cSCC than in AK [21].

2.5. Major Driver Pathways of cSCC

Despite the high mutational load of cSCC and the challenges in determining driver genes within cSCC, many of these genes have been identified, as seen in Table 1. The well-known cSCC drivers are also famously mutated in many other cancers; these include TP53, NOTCH, CDKN2A, PIK3CA, FAT1, RAS, EGFR, and HRAS [7,8,21,23,31,33,34]. These genes often interact with each other in pathways strongly activated in cSCC, such as the PI3/AKT/mTOR, MAPK/AKT, and RAS/RAF/MEK/ERK pathways, and their interactions can be seen in Figure 1 [7,28,29,31]. While many of these driver genes play a role in tumor suppression, and the interplay of their mutations additively contributes to tumorigenesis, the exact mechanisms through which they contribute to malignancy are not fully understood.

2.5.1. TP53

TP53 is one of the most frequently mutated driver genes in cSCC, as well as in almost all human cancers [10,21,27,51]. Its protein product p53 is a tumor suppressor protein that regulates cell cycle arrest, genome stability, DNA damage repair, senescence, and apoptosis [52,53]. Within cSCC, TP53 mutations are very heterogenous, including point mutations, missense mutations, and nonsense mutations that can result in either gains in or a loss of function of the p53 protein [54]. Some TP53 hotspots have been reported in cSCC, at R248W and R248Q, which are conserved arginine locations at the DNA binding interface [54,55,56]. TP53 mutations are suggested to be the initiating mutations in UV-induced cSCC, occurring in 54–95% of cases [7,57]. TP53 interacts with other well-known cSCC driver genes such as CDKN2A, NOTCH1, and NOTCH2, and its mutation has been linked to UV exposure and C > T/CC > TT UV mutational signatures [10,57,58]. Despite its definite role in cSCC tumorigenesis, TP53 mutation alone has not been proven to predict malignancy [19,59]. While many cases of AK have mutations in the TP53 gene, most never progress into cSCCs, making it challenging to consider TP53 a predictive biomarker [19,25]. In addition, TP53 is also mutated in normal skin, meaning that TP53 alone cannot lead to cSCC development, and its interplay with other driver genes must be explored further [15,57].

2.5.2. NOTCH Family

The NOTCH family comprises tumor suppressor genes, particularly NOTCH1, NOTCH2, and NOTCH3, which are mutated in cSCC and encode receptors essential for cell differentiation and cell cycle arrest [60,61,62]. NOTCH1 is the most common driver gene mutated in normal skin, affecting 20% of skin cells [15]. In cSCC, NOTCH1 and NOTCH2 mutations are loss-of-function mutations that occur in around 50–80% of cases [1,5,27,62]. Although NOTCH1 and NOTCH2 are paralogs and present within the same signaling pathway, they are present in different layers of the epidermis, with their differing signals resulting in different cancer prognoses [60,63,64,65,66]. NOTCH1 expression is mediated by p53, and like TP53 mutations, NOTCH1 mutations are believed to occur early on during cSCC [15,60,61,62].

2.5.3. CDKN2A

CDKN2A, or cyclin-dependent kinase inhibitor 2A, is a protein-coding gene that has functionality in G1 cell cycle control [67,68,69]. CDKN2A encodes two proteins, p16INK4a and p14ARF, which are negative regulators of cell proliferation and interact with p53 as part of the cell regulation pathway [60,70]. Like TP53, CDKN2A mutations are present early on in cSCC tumorigenesis [68,70]. In cases of other squamous cell carcinomas, such as head and neck squamous cell carcinoma (HNSCC), patients with severe mutations in both TP53 and CDKN2A have a decreased survival rate and more aggressive cancer forms, suggesting that CDKN2A plays a role in suppressing metastasis [68,70]. This finding was also mirrored in cSCCs, suggesting that CDKN2A mutational presence in a metastatic tumor can predict the prognosis of metastatic cSCC and SCCs [68,70].

2.5.4. PIK3CA Pathway—AKT, mTOR, MAPK, PTEN

The PIK3CA gene encodes phosphoinositide 3-kinases, catalytic enzymes that play a role in the PI3K/AKT/mTOR pathway [29]. The PI3K/AKT/mTOR pathway is consistently activated in cSCC but not in its precursor, AK, suggesting that its dysregulation may be responsible for pre-cancer to cSCC transition [7,28,29,31]. Dysregulation of the pathway can occur at various points: when activated by PI3KCA mutations, upon UV exposure, or after a PTEN loss of function mutation [7,28,29]. PTEN, a tumor suppressor phosphatase, negatively regulates the PI3K/AKT/mTOR pathway by inhibiting PI3K from converting PIP2 into PIP3 [7,29,71]. PTEN has been reported to be mutated in cases of cSCC [5,27,31].
Once PIP2 converts into PIP3, it activates AKT, a serine/threonine protein kinase that regulates terminal differentiation, cell proliferation, the cell cycle, and cell migration through its signaling [7,29,71]. AKT has multiple isoforms that perform independent functions: Akt1, Akt2, and Akt3 [29]. Akt1 is expressed in the top layers of the epidermis and reinforces the epidermal barrier [29,72]. Akt2 is found in the lower layers, is responsible for wound healing and proliferation, and is suggested to contribute more to cSCC tumor progression [29,72,73]. Studies on the two isotopes have inferred that during cSCC progression, Akt1 expression is downregulated while Akt2 expression is upregulated [72]. AKT also plays a critical role in regulation through its intermediate position in the PI3K/AKT/mTOR pathway [7,29,71].
mTOR activation allows for elevated levels of proteins, nucleotides, and lipids, making the protein kinase imperative in sustaining tumor growth [74]. Its position in the PI3K/AKT/mTOR pathway has made it a potential therapeutic target, and mTOR inhibitors, such as rapamycin, have been used to treat a variety of conditions and various other non-cSCC cancers [75].

2.5.5. MAPK(ERK)—RAS/RAF/MEK/ERK Pathway

MAPKs, or mitogen-activated protein kinases, also known as extracellular signal regulated kinases (ERKs), are responsible for signaling roles in many cellular functions, such as cell proliferation, survival, apoptosis, migration, differentiation, and cell cycle regulation [28,29,30,32]. MAPK or ERK is the final factor in the RAS/RAF/MEK/ERK pathway, and is commonly dysregulated across varying cancers [76]. Many studies have found MAPK pathways being activated after UV irradiation and significantly activated in cSCC as opposed to AK, suggesting that the MAPK pathway is critical in the transition from AK to cSCC, possibly through its role in the apoptotic response [28,29,30,32]. Thus, MAPK has been made an ideal target for therapeutic approaches, with studies showing its inhibition in response to photodynamic therapy [32].

2.5.6. RAS Genes: HRAS, NRAS, and KRAS

RAS genes comprise a family of GTPase proteins of three gene isoforms: HRAS, NRAS, and KRAS [76]. Among the three, HRAS is most frequently mutated and associated with cSCC, followed by KRAS, and then in a few cases NRAS [1,5,7,36]. RAS GTPases are upstream of many important pathways in cellular functions, including the PIK3CA and the RAF/MEK/ERK pathways [28]. Upstream of the RAS genes are various receptor tyrosine kinases, one of which is EGFR [77]. When both RAS and EGFR are mutated, there is considerable dysregulation, and the use of EGFR inhibitors becomes ineffective [77]. RAS itself has been proposed as a potential therapeutic target in cSCC [78]. Using salirasib, an anti-RAS drug, L.Li et al., aimed to suppress RAS/RAF/MEK/ERK signaling and in turn decrease the expression of cellular proliferation and regulatory processes; however, the anti-RAS drug was only partially successful in inhibiting the pathway in cell culture [78].

2.5.7. EGFR

EGFR, an epidermal growth factor receptor, is involved in cellular processes such as tissue regeneration, construction, homeostasis, cellular apoptosis, proliferation, and differentiation [28,35,79]. In skin keratinocytes, EGFR is highly mitogenic and activated by UV exposure [79]. In an experiment with EGFR knockout and wild-type mice, El-Abaseri et al., found that after UV exposure, wild-type mice showed cell proliferation that had increased to levels 3× higher than in those in EGFR knockout mice, and that UV-exposed EGFR knockout mice expressed higher levels of apoptosis [79].
Mutations in EGFR resulting in upregulation can be rather disruptive as they have the potential to trickle down to the downstream pathways, further encouraging tumor progression [35]. Within cSCC, EGFR has been reported to have both activating mutations and aberrant overexpression [31,35]. Some studies have found the high epidermal expression of EGFR to be associated with poor prognosis due to increased metastasis and migration to the lymph node [8,35,80,81]. However, other research disputes EGFRs’ correlation with poor outcomes in cSCC, leaving its exact effect on prognosis not understood [80,82,83]. EGFR is upstream of many pathways such as the RAS/RAF/MEK/ERK, PI3K/AKT/mTOR, and JAK/STAT pathways, making its activation very influential and an ideal target for therapeutic interventions [7,28,31,35]. Currently, EGFR inhibitors are available as therapeutic options for select cSCC patients [82].

2.5.8. FAT1

Traditionally downregulated due to a loss of function mutation, FAT1 is a protocadherin known to contribute to cellular migration, mitochondrial respiration, and cell proliferation in normal tissues [27,84,85,86]. In cancer, the FAT1 mutation promotes metastasis, invasiveness, malignant progression, and tumor initiation [1,27,84,85,86]. FAT1 mutations are also found in normal skin but at a much lower rate than other driver genes are found, in only around 3–5% of the skin cells studied [33]. In cSCC models, it was found that FAT1 deletions promote tumor initiation and progression, and increase YAP1 expression, promoting a mesenchymal state within the tumor tissue [86]. In other SCCs, such as esophageal squamous cell carcinoma (ESCC) and hepatocellular carcinoma (HCC), FAT1 was found to be a negative regulator of the epithelial–mesenchymal transition (EMT), with the loss of function of FAT1 promoting EMT, cell growth, migration, and invasion [86,87,88]. This suggests a mechanism in which FAT1 mutations promote tumorigenesis in cSCC and other cancers, but still, more research is needed to investigate this further [86,89].

2.6. Non-Coding Portion of Genome

Mutations in cSCC have also been detected in the non-coding portion of the genome, including telomeric regions of genes [90,91]. Encoded by the hTERT gene, telomerase prevents telomere shortening, but when mutated, telomerase can permit cells to replicate indefinitely and contribute to carcinogenesis [91]. The mutations are present in the TERT promoter region rather than in the hTERT gene sequence [7,44,90]. In cSCC, TERT promoter mutations occur in up to 30–70% of cSCC tumors [7,8,44,91,92,93]. Interestingly, telomere shortening has been reported at almost all stages of cSCC progression, with the telomere length in cSCC found to be significantly shorter than that in its precursor AK [91,92]. According to Campos et al., invasive cSCCs have more mutations in the TERT promoter than non-invasive cSCCs do, and these mutations can independently predict the local recurrence of cSCC [44].
Other alterations within non-coding portions of the genome that have been reported in cSCC include long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), and microRNAs (miRNAs) [94,95]. Long non-coding RNAs (lncRNAs), regulatory RNAs with over 200 nucleotides, and circular RNAs (circRNAs), RNA back-spliced into a continuous loop, have been identified in cSCC as novel regulators of gene expression [94]. Mahapatra et al., detected 908 lncRNAs and 55 circRNAs to be differentially expressed in cSCC [94]. Among these are known oncogenic lncRNAs: PVT1, LUCAT1, and CASC9 [94]. Novel skin-specific lncRNAs were also found to be dysregulated [94]. A global reduction in circRNA was seen in cSCC as opposed to normal skin [94].
Many miRNAs have been reported to be dysregulated in cSCC, including the upregulation of Onco-miRNAs miRNA-21, miRNA-31, miRNA-135b, and miRNA-365 [96]. Interestingly, many miRNA target genes are within the known cSCC pathways, mediating genes like PTEN, KRAS, and ERK [97,98,99,100]. Specifically, PTEN is a frequent target of many miRNAs types such as 142, 221, and 21 [95,97,101,102].

2.7. Novel cSCC Driver Genes

In the last ten years, increased efforts to genotype and analyze the mutational landscape of metastatic, aggressive, advanced, and traditional cSCC have allowed for various potential driver genes to be discovered. Table 2 lists genes that have been recognized among several primary research papers as relevantly mutated and/or nominated as novel driver genes across the cSCC subtypes. The following genes were nominated in three primary research papers: CCND1, CREBBP, EP300, ERBB4, EZH2, FGFR3, KMT2C, KMT2D, and TGFBR2 [1,5,31,37,38,39,40,41,42,50]. Additionally, genes that were nominated in two primary sources include AJUBA, ARID2, ATM, BRAF, BRCA2, CASP8, KNSTRN, LRP1B, NFE2L2, CARD11, MYC, and RIPK4 [1,5,31,37,39,40,42,43,103,104]. Of the genes nominated, many are known contributors in other cancers [1,31]. The genes RIPK4, CCND1, CASP8, and MYC are also reported to be commonly mutated or dysregulated in HNSCC [31]. BRAF mutations, found at very low occurrence rates in cSCC, are linked with several different cancers, such as metastatic colorectal cancer, thyroid cancer, and melanoma [31,105,106].
While there are many limitations in uncovering these novel drivers, the genetic analysis and mutation rates of these drivers are crucial in understanding cSCC development, and in turn can aid in finding new therapeutic drug targets.

3. Tumor Microenvironment

As previously mentioned, there is still a lack of clear understanding in the process of the malignant transformation of cSCC, as mutational landscapes can be similar between normal skin, pre-cancers, and invasive cSCC. Therefore, understanding other external factors such as the tumor microenvironment is critical to elucidating mechanisms of cSCC pathogenesis. As solid organ transplant recipients (OTRs) are at a 50–100-fold increased risk for the development of cSCC, their immunosuppressed microenvironment is a clear contributor to cSCC pathogenesis [122,123,124,125]. Generally, OTRs have decreased amounts of immune infiltrates, including lower B-cell infiltrates, as well as a variety of T-cell types (CD4+, CD4+T helper 1, CD8+, cytotoxic CD8+, Naive CD8+, etc.) [122,123,124,125]. While the risk for skin cancers is most significant, it is understood that OTR patients are also at an increased risk of developing other cancers, such as Non-Hodgkin’s lymphoma, kidney cancer, liver cancer, and Kaposi’s sarcoma [126,127]. Therefore, herein, we also explore the current understanding of tumorigenesis beyond the genome, focusing on the extracellular matrix, stromal cells, and immune cells [128,129].

3.1. The Extracellular Matrix and MMPs

The extracellular matrix (ECM) is a critical cell microenvironment component. While non-cellular itself, it provides cells with structure and an opportunity to interact with each other through extracellular signaling [130]. During cSCC progression, ECM remodeling occurs, often forming a dense fibrosis around solid tumors [131,132,133]. These modifications can be attributed to proteolytic enzymes, such as matrix metalloproteinases (MMPs) and A disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS) [130,134,135]. With their ability to break down proteins, MMPs and ADAMTS can completely denature and reorganize the ECM to facilitate the development of cSCC [134,135].

3.1.1. MMPs

MMPs are found to be upregulated in cSCC, and their increased expression is deemed a significant biomarker in predicting prognosis, aggressiveness, and metastasis [130,133,134]. MMP-13 expression has been proposed to detect the level of invasiveness of cSCC and can be used to monitor progression [133]. MMP-9 and MMP-11 expression encourages matrix turnover, while MMP-10 enhances this process and angiogenesis, facilitating metastasis in cSCC [134,136,137,138]. Additionally, MMP-7 expression was found in the invasive front of cSCC tumors, suggesting a role in tumor progression [139].

3.1.2. Collagen

Collagen is a fibrous protein that makes up the majority of the ECM [130,134]. The 28 types of collagens identified to date are versatile and abundant within the skin, making them candidates for cancer complicity [140]. In cSCC, collagen XVII is expressed widely by tumor cells at high expression levels not seen in normal skin or cSCC precursors [141]. Another collagen, collagen VII, suppresses TGFβ signaling and angiogenesis in cSCC [142]. In vivo, the knockout of collagen VII promoted angiogenesis, with collagen VII treatment decreasing tumor angiogenesis [142]. Less is known about collagen XV’s contribution to cSCC tumorigenesis, but it is highly expressed in cSCC tumors and particularly abundant in tumor stromal cells, suggesting that collagen XV may be linked to cancer-associated fibroblasts [142].

3.1.3. Stromal Cells

Stromal cells normally aid in the regeneration of tissues, immune responses, ECM remodeling, and tissue homeostasis [143,144]. In the tumor microenvironment, stromal cells known as cancer-associated fibroblasts (CAFs) promote tumorigenesis by providing growth factors and nutrients to the newly formed tumor and blood vessels [145]. In cSCC specifically, Martínez-Nieto et al., found that integrin α11β1 was upregulated in cancer-associated fibroblasts and other human neoplasms [129]. Additionally, α11-deficient cells hindered stromal cells’ ability to differentiate into tumor-promoting CAFs, and as a result, the mice experimented on had decreased tumor cell proliferation, development, and burden [129]. This suggests that α11 expression in stromal cells helps cSCC tumors to progress to malignant states [129]. Studies have also found that the secretion of growth factor TGFβ1 by CAFs contributes to increased resistance to photodynamic therapy, signifying that CAF-secreted TGFβ1 can be used as a biomarker to determine the effectiveness of photodynamic therapy in cSCC patients [144].

3.2. Immune Cell Populations

3.2.1. Macrophages

In normal tissues, macrophages are an integral part of the immune system, removing threats to the immune system through phagocytosis [146]. In cancer tissue, tumor-associated macrophages (TAMs) can alter both the tumor and immune microenvironment [138].
TAMs are more abundant in cSCC than normal skin, and studies show that these TAMs promote tumor growth and metastasis [138,147]. TAMs can produce VEGF-C, vascular endothelial growth factor to stimulate growth, lymph angiogenesis, and metastasis [138,148,149]. They also contribute to cSCC tumorigenesis through their generation and secretion of MMPs, specifically MMP9 and MMP11 [138].
Within the cSCC microenvironment, TAMs are in heterogeneous activation states, either M1 or M2, with some being bi-activated, expressing both M1 and M2 markers [138]. TAMs under M2 activation tend to promote tumorigenesis, while M1 macrophages promote the pro-inflammatory response and induce the anti-tumor immune response [148,150]. Notably, OTR patients demonstrate similar levels of TAM infiltration but significantly fewer M2-activated TAMs [151]. While more recent research suggests that TAMs are not strictly adherent to M1/M2 states and can be alternatively defined based on their effect on the TME as either pro- or anti-inflammatory [152], most studies on cSCC have focused on traditional M1/M2 classifications.
In general, an increase in TAMs, especially M2 ones, is associated with poor prognosis across many other cancers, such as breast cancer, oral cutaneous cell carcinoma, and prostate [138,153,154,155,156,157,158]. TAMs are a potential target in immunotherapy, as the repolarization of TAMs into anti-tumor macrophages could be used to slow tumor progression [159].

3.2.2. T-Cells

Limited studies have examined T-cell populations within cSCC, focusing mainly on the immune microenvironment differences between cSCC and OTR cSCC. The latter is generally more aggressive, implying that microenvironmental differences across immune states could explain the variation in responses to immunotherapy.
Cytotoxic T-cells from OTR patients respond differently than those in regular cSCC patients, favoring the production of cytokine IL-22 [160]. Both cSCC and OTR cSCC show significantly higher numbers of CD3+, T-Lymphocytes, and CD8+, cytotoxic T-cells, as opposed to normal skin, with cSCC having significantly more of these cells than OTR cSCC [122,160]. Topographically, in both cSCC and OTR cSCC, CD3+ and CD8+ T-cells are located primarily in peritumoral regions as opposed to within the tumor nests [160].
In cancer, T-regulatory (Treg) cells can suppress anti-tumor responses, with higher proportions of tumor-infiltrating Treg cells correlating with poorer prognosis [161,162,163,164,165]. This pattern has also been noted in cSCC [165,166]. Tregs are more abundant in cSCC sites than in the peripheral blood, contributing to metastasis through their suppression of effector T-cells and IFN-γ expression [165]. While Treg cells are reported at similar levels in cSCC and OTR cSCC, both are at significantly higher levels than in normal skin [122,160]. The Treg-to-cytotoxic T-cell ratio is higher in OTR cSCC than in cSCC, potentially promoting immune evasion [122,160].
cSCC patients had higher levels of naïve T-cells than OTR cSCC patients did, but shared similar numbers of exhausted T-cells [122]. Within the same patient, high intertumoral heterogeneity of T-cell populations can be present [122]. One report showed that, in a single OTR patient, a forehead tumor demonstrated mainly cytotoxic T-cells, while a tumor from the neck predominantly showed exhausted T-cells [122]. In general, exhausted and effector T-cells had the highest proliferation capacity across the T-cell subsets, with exhausted CD4+ and CD8+ cells expressing inhibitory receptors and exhaustion markers [167].

3.2.3. Natural Killer Cells

Natural killer (NK) cells, as part of the innate immune system, can kill tumor cells without antigen priming or major histocompatibility complex (MHC) interactions, and without harming normal cells [168]. Generally, in cancer, NK cell functions are diminished [168,169]. Higher rates of cSCC are seen in patients with functional NK cell deficiencies, implying that NK cells play a role in inhibiting cSCC progression [170,171].
Studies using Nfil3−/− knockout mice demonstrated NK cells’ critical role in early cSCC tumorigenesis, as these mice developed significantly more papillomas and pre-cancerous protrusions, and had increased tumor growth [169]. When these tumor-bearing mice were treated with NK cells, the tumor size decreased to levels that were near those of wild-type Nfil3+/+ [169]. However, NK cells isolated from these tumors showed reduced cytotoxicity, IFN-γ production, and secretion [169].
NK cells’ pivotal role and dysfunction in the TME has prompted research into their therapeutic potential. A recent preclinical model explored if the transplantation of NK cells from healthy donors into non-autologous patients could serve as a potential therapeutic approach [168]. In vitro, the co-incubation of SCC-13 cells with healthy NK cells resulted in a dose-dependent reduction in the size of spheroids and in the number of tumor cells [168]. In vivo, mice with SCC-13 xenograft tumors treated with NK cells saw a 70% reduction in tumor volume at three weeks [168]. In both in vivo and in vitro models, a reduction in YAP1 and MEK1/2-P levels occurred, demonstrating that the healthy NK cells were able to induce apoptosis [168].

3.2.4. Neutrophils

In vivo and in vitro experiments have found that tumor-associated neutrophils (TANs) are present in both pre-cancerous and cancerous cSCC lesions, with 30–80% of neutrophils present exhibiting the TAN phenotype [172]. In general, neutrophils were localized to the pre-cancerous and cancerous regions as opposed to the surrounding skin, with significantly higher percentages in tumors as opposed to the papillomas [172]. The orthotopic implantation of cell line mSCC38 in mice also supported these findings, with neutrophil frequency being positively correlated to the mSCC38 tumor volume and having decreased frequency in the surrounding skin [172].
The functions of TANs evolve with the carcinogenesis stages. In pre-cancerous stages they contribute to ECM remodeling and angiogenesis [172]. In cancerous stages, TANs actively suppress immune responses by inhibiting CD8+ T-cell responses and upregulating PD-L1 [172]. In vivo depletion of neutrophils significantly increased the proliferation of CD8+ T-cells and elevated IFN-γ levels, further supporting the immunosuppressive role of TANs in cSCC [172].
Clinically, elevated neutrophil counts have been associated with increased tumor thickness and poor overall survival in cSCC patients, warranting further investigation of neutrophils as a potential prognostic marker [173].

4. Novel Therapeutics

With the current understanding of driver genes of cSCC and the tumor microenvironment in cSCC, certain therapeutic approaches have been pursued. In recent years, novel therapies, including immune checkpoint inhibitors and EGFR inhibitors, have demonstrated promising results for the treatment of cSCC, particularly in unresectable or recurrent/metastatic disease (Table 3). While overall response rates (ORRs) to these treatments can range between 8% and 63%, patients who do not respond and OTR patients who cannot receive immune checkpoint inhibitors are in need of novel therapies.

4.1. Immunotherapy

Immunotherapy has revolutionized cancer treatment in the past decade. These therapies work by upregulating immune activation to produce an anti-tumor response, specifically through monoclonal antibodies inhibiting immune checkpoints CTLA-4 or PD-1/PD-L1 [182]. In the last 6 years, two anti-PD1 immunotherapy drugs have been approved by the FDA for cSCC treatment: cemiplimab and pembrolizumab [11,183,184].
In 2018, cemiplimab became the first FDA-approved PD-1 checkpoint immune inhibitor for metastatic or local advanced cSCC [185]. This approval stemmed from phase I and phase II clinical trials conducted by Migden et al., which demonstrated durable and promising responses in about half of the patients [11]. In a phase II trial, 79 advanced cSCC patients not amenable to surgery or radiation were treated with cemiplimab [12]. This showed a high disease control rate of 79% in patients with at least stable disease, with 31% showing partial response and 13% showing complete response [12]. Following the FDA approval of cemiplimab in 2018, trials with pembrolizumab demonstrated similar efficacy in cSCC, leading to subsequent approval in 2020 [83,174]. Subsequently, a recent phase II neoadjuvant study for previously untreated resectable cSCC demonstrated that administering 2-4 cycles of cemiplimab before surgery led to the achievement of a complete pathologic response in 51% of patients and a major pathologic response in 63% (<10% viable tumor) [12]. Given these high pathologic response rates, future studies are investigating the long-term impact of neoadjuvant treatment as well as the role of adjuvant immunotherapy after surgery. In melanoma, the addition of neoadjuvant immunotherapy before surgery significantly improved outcomes compared to using immunotherapy in the adjuvant setting after surgery, demonstrating the potential advantage of the early introduction of these treatments before the removal of the primary tumor [186].

4.2. EGFR Inhibitors

When patients are elderly, or immunocompromised, and surgery and radiation therapy fail, the use of EGFR inhibitors is another treatment option for cSCC. As mentioned previously, EGFR dysregulation is a known characteristic within cSCC; however, the exact source of EGFR dysregulation is an important determinant in the success of EGFR inhibitor therapy. While EGFR dysregulation is found in cSCC patients, downstream RAS mutations can limit drug efficacy [31,82,83]. Multiple agents targeting EGFR are available, including monoclonal antibodies such as cetuximab and panitumumab, and small-molecule inhibitors such as gefitinib and erlotinib. The effectiveness of these inhibitors in cSCC are being investigated, both as a monotherapy and in addition to radiation or immunotherapy [83,187,188].

4.2.1. EGFR Monotherapy

Usually, when immunotherapy is not an option for patients, EGFR monotherapy can be considered. In a 2011 phase II clinical study by Maubec et al., 36 patients with unresectable cSCC received cetuximab as a monotherapy [175]. At 6 weeks, 68% of the patients were able to achieve stable disease [175]. However, only 6% achieved a CR, 26% had a PR, and 19% had progressed disease [175]. In a retrospective analysis conducted by Marin-Acevedo et al., cetuximab showed a 64% ORR (n = 11) in patients after failing anti-PD1 therapy, and an 80% ORR (n = 10) in anti-PD1-naïve patients [187]. While promising, it is important to note that retrospective studies are limited by their selection bias, small sample size, and heterogenous population.
Panitumumab has also shown promise as a monotherapy for cSCC. In the phase II (n = 16) study conducted by Foote et al., it was found that patients with locally advanced, metastatic, or recurrent cSCC, previously treated with either radiotherapy (n = 14) and/or cytotoxic chemotherapy (n = 7), saw an overall response rate of 31%, with 6 of the 16 successfully achieving stable disease [176]. Phase II clinical trials have also been conducted to investigate the effect of erlotinib and gefitinib on cSCC; however, they were only reported to have modest responses [177,178]. It is important to note that these studies are limited by their sample size; thus, more research is needed to further confirm the positive results.

4.2.2. EGFR Inhibitors with Radiation

Currently, there have only been retrospective studies on the efficacy of EGFR and radiation combination therapy for cSCC. Most recently, a retrospective case series of 18 patients with locally advanced cSCC found a 55.5% CR in combinatory radiation and cetuximab therapy [189]. Nevertheless, high ORR in other SCCs, such as HNSCC, suggest that this combinatory therapy could also be effective in cSCC. In locally advanced HNSCC, Bonner et al., saw improved overall survival when cetuximab was combined with radiation (a median survival of 49 months) compared to that when using radiotherapy alone (29.3 months) [190].

4.2.3. EGFR Inhibitors with Immunotherapy

Combinatory therapy with cetuximab and anti-PD1 inhibitors still needs to be explored further, with retrospective and case reports suggesting promise in its efficacy. In Hsu et al., panitumumab was used in combination with anti-PD1 in three patients who had progressed on anti-PD1 inhibitors, with all patients achieving durable CR [191].
While limited data exist on the combination of EGFR inhibitors with immunotherapy in cSCC, the use of this combinatory treatment in other squamous cell carcinomas suggests that it could also be successful in cSCC [180,181]. Sacco et al., conducted a phase II trial treating recurrent or metastatic HNSCC with cetuximab followed by pembrolizumab [180]. After 6 months, there was an ORR of 45% [180]. In another phase II trial treating recurrent/metastatic HNSCC patients with cetuximab followed by nivolumab, Chung et al., found an ORR of 23% in patients with prior treatment for recurrent/metastatic disease and 33% in patients for which therapy was first line for recurrent/metastatic disease [181].

5. Conclusions

The high mutational burden in cSCC complicates our understanding of its biological underpinnings. This complexity makes it challenging to differentiate between passenger alterations present in normal skin and premalignant lesions, and to identify the driver alterations that contribute to carcinogenesis. The tumor microenvironment also contributes significantly to tumor progression, and the interplay between the immune system has allowed for the success of immunotherapies in the treatment of cSCC. For patients who do not respond to immunotherapy, novel targeted treatments are under investigation in targeting pathways including EGFR inhibition. Additional therapeutic opportunities may be available in targeting other pathways such as mTOR inhibitors, photodynamic therapy, or cancer-associated fibroblasts and tumor-associated macrophages. Continued research integrating genomic profiling, microenvironment analysis, and novel therapeutic approaches holds promise for improving outcomes in cSCC patients.

Author Contributions

Conceptualization, T.M.H. and T.G.; methodology, writing—original draft preparation, T.M.H.; writing—review and editing, T.M.H., T.G. and S.J.P.; visualization, T.M.H.; supervision, T.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Gleiberman Head & Neck Cancer Excellence Fund and The APC was funded by Gleiberman Head & Neck Cancer Excellence Fund.

Acknowledgments

Figure 1 was created with Biorender.com (accessed on 15 July 2024).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Pickering, C.R.; Zhou, J.H.; Lee, J.J.; Drummond, J.A.; Peng, S.A.; Saade, R.E.; Tsai, K.Y.; Curry, J.L.; Tetzlaff, M.T.; Lai, S.Y.; et al. Mutational Landscape of Aggressive Cutaneous Squamous Cell Carcinoma. Clin. Cancer Res. 2014, 20, 6582–6592. [Google Scholar] [CrossRef] [PubMed]
  2. Que, S.K.T.; Zwald, F.O.; Schmults, C.D. Cutaneous Squamous Cell Carcinoma: Incidence, Risk Factors, Diagnosis, and Staging. J. Am. Acad. Dermatol. 2018, 78, 237–247. [Google Scholar] [CrossRef]
  3. Bradford, P.T.; Goldstein, A.M.; Tamura, D.; Khan, S.G.; Ueda, T.; Boyle, J.; Oh, K.-S.; Imoto, K.; Inui, H.; Moriwaki, S.-I.; et al. Cancer and neurologic degeneration in xeroderma pigmentosum: Long term follow-up characterizes the role of dna repair. J. Med. Genet. 2011, 48, 168–176. [Google Scholar] [CrossRef]
  4. Burton, K.A.; Ashack, K.A.; Khachemoune, A. Cutaneous Squamous Cell Carcinoma: A Review of High-Risk and Metastatic Disease. Am. J. Clin. Dermatol. 2016, 17, 491–508. [Google Scholar] [CrossRef]
  5. Chang, D.; Shain, A.H. The Landscape of Driver Mutations in Cutaneous Squamous Cell Carcinoma. NPJ Genom. Med. 2021, 6, 61. [Google Scholar] [CrossRef]
  6. Combalia, A.; Carrera, C. Squamous Cell Carcinoma: An Update on Diagnosis and Treatment. Dermatol. Pract. Concept. 2020, 10, e2020066. [Google Scholar] [CrossRef]
  7. Di Nardo, L.; Pellegrini, C.; Di Stefani, A.; Del Regno, L.; Sollena, P.; Piccerillo, A.; Longo, C.; Garbe, C.; Fargnoli, M.C.; Peris, K. Molecular Genetics of Cutaneous Squamous Cell Carcinoma: Perspective for Treatment Strategies. J. Eur. Acad. Dermatol. Venereol. 2020, 34, 932–941. [Google Scholar] [CrossRef] [PubMed]
  8. Griewank, K.G.; Murali, R.; Schilling, B.; Schimming, T.; Möller, I.; Moll, I.; Schwamborn, M.; Sucker, A.; Zimmer, L.; Schadendorf, D.; et al. TERT Promoter Mutations Are Frequent in Cutaneous Basal Cell Carcinoma and Squamous Cell Carcinoma. PLoS ONE 2013, 8, e80354. [Google Scholar] [CrossRef]
  9. Fania, L.; Didona, D.; Di Pietro, F.R.; Verkhovskaia, S.; Morese, R.; Paolino, G.; Donati, M.; Ricci, F.; Coco, V.; Ricci, F.; et al. Cutaneous Squamous Cell Carcinoma: From Pathophysiology to Novel Therapeutic Approaches. Biomedicines 2021, 9, 171. [Google Scholar] [CrossRef]
  10. Wikonkal, N.M.; Brash, D.E. Ultraviolet Radiation Induced Signature Mutations in Photocarcinogenesis. J. Investig. Dermatol. Symp. Proc. 1999, 4, 6–10. [Google Scholar] [CrossRef] [PubMed]
  11. 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]
  12. Gross, N.D.; Miller, D.M.; Khushalani, N.I.; Divi, V.; Ruiz, E.S.; Lipson, E.J.; Meier, F.; Su, Y.B.; Swiecicki, P.L.; Atlas, J.; et al. Neoadjuvant Cemiplimab for Stage II to IV Cutaneous Squamous-Cell Carcinoma. N. Engl. J. Med. 2022, 387, 1557–1568. [Google Scholar] [CrossRef]
  13. Pfeifer, G.P. Mechanisms of UV-Induced Mutations and Skin Cancer. Genome Instab. Dis. 2020, 1, 99–113. [Google Scholar] [CrossRef] [PubMed]
  14. Brash, D.E. UV Signature Mutations. Photochem. Photobiol. 2015, 91, 15–26. [Google Scholar] [CrossRef]
  15. Martincorena, I.; Roshan, A.; Gerstung, M.; Ellis, P.; Van Loo, P.; McLaren, S.; Wedge, D.C.; Fullam, A.; Alexandrov, L.B.; Tubio, J.M.; et al. High Burden and Pervasive Positive Selection of Somatic Mutations in Normal Human Skin. Science 2015, 348, 880–886. [Google Scholar] [CrossRef]
  16. Shen, Y.; Ha, W.; Zeng, W.; Queen, D.; Liu, L. Exome Sequencing Identifies Novel Mutation Signatures of UV Radiation and Trichostatin A in Primary Human Keratinocytes. Sci. Rep. 2020, 10, 4943. [Google Scholar] [CrossRef]
  17. Brambullo, T.; Colonna, M.R.; Vindigni, V.; Piaserico, S.; Masciopinto, G.; Galeano, M.; Costa, A.L.; Bassetto, F. Xeroderma Pigmentosum: A Genetic Condition Skin Cancer Correlated—A Systematic Review. BioMed Res. Int. 2022, 2022, 8549532. [Google Scholar] [CrossRef]
  18. DiGiovanna, J.J.; Kraemer, K.H. Shining a Light on Xeroderma Pigmentosum. J. Investig. Dermatol. 2012, 132, 785–796. [Google Scholar] [CrossRef]
  19. Neto, P.D.; Alchorne, M.; Michalany, N.; Abreu, M.; Borra, R. Reduced P53 Staining in Actinic Keratosis Is Associated with Squamous Cell Carcinoma: A Preliminary Study. Indian J. Dermatol. 2013, 58, 325. [Google Scholar] [CrossRef]
  20. Reinehr, C.P.H.; Bakos, R.M. Actinic Keratoses: Review of Clinical, Dermoscopic, and Therapeutic Aspects. Bras. Dermatol. 2019, 94, 637–657. [Google Scholar] [CrossRef]
  21. Thomson, J.; Bewicke-Copley, F.; Anene, C.A.; Gulati, A.; Nagano, A.; Purdie, K.; Inman, G.J.; Proby, C.M.; Leigh, I.M.; Harwood, C.A.; et al. The Genomic Landscape of Actinic Keratosis. J. Investig. Dermatol. 2021, 141, 1664–1674.e7. [Google Scholar] [CrossRef]
  22. Campbell, C.; Quinn, A.G.; Ro, Y.-S.; Angus, B.; Rees, J.L. P53 Mutations Are Common and Early Events That Precede Tumor Invasion in Squamous Cell Neoplasia of the Skin. J. Investig. Dermatol. 1993, 100, 746–748. [Google Scholar] [CrossRef]
  23. Nissinen, L.; Farshchian, M.; Riihilä, P.; Kähäri, V.-M. New Perspectives on Role of Tumor Microenvironment in Progression of Cutaneous Squamous Cell Carcinoma. Cell Tissue Res. 2016, 365, 691–702. [Google Scholar] [CrossRef]
  24. Wei, L.; Christensen, S.R.; Fitzgerald, M.E.; Graham, J.; Hutson, N.D.; Zhang, C.; Huang, Z.; Hu, Q.; Zhan, F.; Xie, J.; et al. Ultradeep Sequencing Differentiates Patterns of Skin Clonal Mutations Associated with Sun-Exposure Status and Skin Cancer Burden. Sci. Adv. 2021, 7, eabd7703. [Google Scholar] [CrossRef]
  25. Criscione, V.D.; Weinstock, M.A.; Naylor, M.F.; Luque, C.; Eide, M.J.; Bingham, S.F.; Department of Veteran Affairs Topical Tretinoin Chemoprevention Trial Group. Actinic Keratoses: Natural History and Risk of Malignant Transformation in the Veterans Affairs Topical Tretinoin Chemoprevention Trial. Cancer 2009, 115, 2523–2530. [Google Scholar] [CrossRef]
  26. Siegel, J.A.; Korgavkar, K.; Weinstock, M.A. Current Perspective on Actinic Keratosis: A Review. Br. J. Dermatol. 2017, 177, 350–358. [Google Scholar] [CrossRef] [PubMed]
  27. Inman, G.J.; Wang, J.; Nagano, A.; Alexandrov, L.B.; Purdie, K.J.; Taylor, R.G.; Sherwood, V.; Thomson, J.; Hogan, S.; Spender, L.C.; et al. The Genomic Landscape of Cutaneous SCC Reveals Drivers and a Novel Azathioprine Associated Mutational Signature. Nat. Commun. 2018, 9, 3667. [Google Scholar] [CrossRef] [PubMed]
  28. Einspahr, J.G.; Calvert, V.; Alberts, D.S.; Curiel-Lewandrowski, C.; Warneke, J.; Krouse, R.; Stratton, S.P.; Liotta, L.; Longo, C.; Pellicani, G.; et al. Functional Protein Pathway Activation Mapping of the Progression of Normal Skin to Squamous Cell Carcinoma. Cancer Prev. Res. 2012, 5, 403–413. [Google Scholar] [CrossRef]
  29. Janus, J.M.; O’Shaughnessy, R.F.L.; Harwood, C.A.; Maffucci, T. Phosphoinositide 3-Kinase-Dependent Signalling Pathways in Cutaneous Squamous Cell Carcinomas. Cancers 2017, 9, 86. [Google Scholar] [CrossRef]
  30. Lambert, S.R.; Mladkova, N.; Gulati, A.; Hamoudi, R.; Purdie, K.; Cerio, R.; Leigh, I.; Proby, C.; Harwood, C.A. Key Differences Identified between Actinic Keratosis and Cutaneous Squamous Cell Carcinoma by Transcriptome Profiling. Br. J. Cancer 2014, 110, 520–529. [Google Scholar] [CrossRef]
  31. Li, Y.Y.; Hanna, G.J.; Laga, A.C.; Haddad, R.I.; Lorch, J.H.; Hammerman, P.S. Genomic Analysis of Metastatic Cutaneous Squamous Cell Carcinoma. Clin. Cancer Res. 2015, 21, 1447–1456. [Google Scholar] [CrossRef] [PubMed]
  32. Zhao, Y.; Dong, J.; Liao, Y.; Wang, H.; Zhou, D.; Kang, J.; Chen, X. Identification and Validation of Four Photodynamic Therapy Related Genes Inhibiting MAPK and Inducing Cell Cycle Alteration in Squamous Cell Carcinoma. Front. Oncol. 2022, 12, 946493. [Google Scholar] [CrossRef]
  33. Martincorena, I.; Fowler, J.C.; Wabik, A.; Lawson, A.R.J.; Abascal, F.; Hall, M.W.J.; Cagan, A.; Murai, K.; Mahbubani, K.; Stratton, M.R.; et al. Somatic Mutant Clones Colonize the Human Esophagus with Age. Science 2018, 362, 911–917. [Google Scholar] [CrossRef]
  34. Zheng, Y.; Chi, S.; Li, C. Identification of Potential Gene Drivers of Cutaneous Squamous Cell Carcinoma. Medicine 2020, 99, e22257. [Google Scholar] [CrossRef]
  35. de Lima, P.O.; Joseph, S.; Panizza, B.; Simpson, F. Epidermal Growth Factor Receptor’s Function in Cutaneous Squamous Cell Carcinoma and Its Role as a Therapeutic Target in the Age of Immunotherapies. Curr. Treat. Options Oncol. 2020, 21, 9. [Google Scholar] [CrossRef]
  36. Bailey, P.; Ridgway, R.A.; Cammareri, P.; Treanor-Taylor, M.; Bailey, U.-M.; Schoenherr, C.; Bone, M.; Schreyer, D.; Purdie, K.; Thomson, J.; et al. Driver Gene Combinations Dictate Cutaneous Squamous Cell Carcinoma Disease Continuum Progression. Nat. Commun. 2023, 14, 5211. [Google Scholar] [CrossRef]
  37. Al-Rohil, R.N.; Tarasen, A.J.; Carlson, J.A.; Wang, K.; Johnson, A.; Yelensky, R.; Lipson, D.; Elvin, J.A.; Vergilio, J.-A.; Ali, S.M.; et al. Evaluation of 122 Advanced-Stage Cutaneous Squamous Cell Carcinomas by Comprehensive Genomic Profiling Opens the Door for New Routes to Targeted Therapies. Cancer 2016, 122, 249–257. [Google Scholar] [CrossRef]
  38. Cammareri, P.; Rose, A.M.; Vincent, D.F.; Wang, J.; Nagano, A.; Libertini, S.; Ridgway, R.A.; Athineos, D.; Coates, P.J.; McHugh, A.; et al. Inactivation of TGFβ Receptors in Stem Cells Drives Cutaneous Squamous Cell Carcinoma. Nat. Commun. 2016, 7, 12493. [Google Scholar] [CrossRef] [PubMed]
  39. Jones, J.; Wetzel, M.; Brown, T.; Jung, J. Molecular Profile of Advanced Cutaneous Squamous Cell Carcinoma. J. Clin. Aesthet. Dermatol. 2021, 14, 32–38. [Google Scholar]
  40. Oberholzer, P.A.; Kee, D.; Dziunycz, P.; Sucker, A.; Kamsukom, N.; Jones, R.; Roden, C.; Chalk, C.J.; Ardlie, K.; Palescandolo, E.; et al. RAS Mutations Are Associated with the Development of Cutaneous Squamous Cell Tumors in Patients Treated with RAF Inhibitors. J. Clin. Oncol. 2012, 30, 316–321. [Google Scholar] [CrossRef]
  41. Ridd, K.; Bastian, B.C. Somatic Mutation of Epidermal Growth Factor Receptor in a Small Subset of Cutaneous Squamous Cell Carcinoma. J. Investig. Dermatol. 2010, 130, 901–903. [Google Scholar] [CrossRef]
  42. Yilmaz, A.S.; Ozer, H.G.; Gillespie, J.; Allain, D.C.; Bernhardt, M.N.; Furlan, K.C.; Castro, L.T.; Peters, S.B.; Nagarajan, P.; Kang, S.; et al. Differential Mutation Frequencies in Metastatic Cutaneous Squamous Cell Carcinomas versus Primary Tumors. Cancer 2017, 123, 1184–1193. [Google Scholar] [CrossRef]
  43. Watt, S.A.; Purdie, K.J.; den Breems, N.Y.; Dimon, M.; Arron, S.T.; McHugh, A.T.; Xue, D.J.; Dayal, J.H.S.; Proby, C.M.; Harwood, C.A.; et al. Novel CARD11 Mutations in Human Cutaneous Squamous Cell Carcinoma Lead to Aberrant NF-κB Regulation. Am. J. Pathol. 2015, 185, 2354–2363. [Google Scholar] [CrossRef] [PubMed]
  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. Mizukami, T.; Izawa, N.; Nakajima, T.E.; Sunakawa, Y. Targeting EGFR and RAS/RAF Signaling in the Treatment of Metastatic Colorectal Cancer: From Current Treatment Strategies to Future Perspectives. Drugs 2019, 79, 633–645. [Google Scholar] [CrossRef]
  46. Hertzler-Schaefer, K.; Mathew, G.; Somani, A.-K.; Tholpady, S.; Kadakia, M.P.; Chen, Y.; Spandau, D.F.; Zhang, X. Pten Loss Induces Autocrine FGF Signaling to Promote Skin Tumorigenesis. Cell Rep. 2014, 6, 818–826. [Google Scholar] [CrossRef]
  47. Arnault, J.P.; Mateus, C.; Escudier, B.; Tomasic, G.; Wechsler, J.; Hollville, E.; Soria, J.-C.; Malka, D.; Sarasin, A.; Larcher, M.; et al. Skin Tumors Induced by Sorafenib; Paradoxic RAS–RAF Pathway Activation and Oncogenic Mutations of HRAS, TP53, and TGFBR1. Clin. Cancer Res. 2012, 18, 263–272. [Google Scholar] [CrossRef]
  48. Pacifico, A.; Goldberg, L.H.; Peris, K.; Chimenti, S.; Leone, G.; Ananthaswamy, H.N. Loss of CDKN2A and p14ARF Expression Occurs Frequently in Human Nonmelanoma Skin Cancers. Br. J. Dermatol. 2008, 158, 291–297. [Google Scholar] [CrossRef] [PubMed]
  49. Campos, M.A.; Macedo, S.; Fernandes, M.S.; Pestana, A.; Pardal, J.; Batista, R.; Vinagre, J.; Sanches, A.; Baptista, A.; Lopes, J.M.; et al. Prognostic Significance of RAS Mutations and P53 Expression in Cutaneous Squamous Cell Carcinomas. Genes 2020, 11, 751. [Google Scholar] [CrossRef] [PubMed]
  50. Rose, A.M.; Sansom, O.J.; Inman, G.J. Loss of TGF-β Signaling Drives cSCC from Skin Stem Cells—More Evidence. Cell Cycle 2016, 16, 386–387. [Google Scholar] [CrossRef]
  51. Olivier, M.; Hollstein, M.; Hainaut, P. TP53 Mutations in Human Cancers: Origins, Consequences, and Clinical Use. Cold Spring Harb. Perspect. Biol. 2010, 2, a001008. [Google Scholar] [CrossRef]
  52. Aubrey, B.J.; Strasser, A.; Kelly, G.L. Tumor-Suppressor Functions of the TP53 Pathway. Cold Spring Harb. Perspect. Med. 2016, 6, a026062. [Google Scholar] [CrossRef] [PubMed]
  53. Ozaki, T.; Nakagawara, A. Role of P53 in Cell Death and Human Cancers. Cancers 2011, 3, 994–1013. [Google Scholar] [CrossRef]
  54. Piipponen, M.; Riihilä, P.; Nissinen, L.; Kähäri, V.-M. The Role of P53 in Progression of Cutaneous Squamous Cell Carcinoma. Cancers 2021, 13, 4507. [Google Scholar] [CrossRef]
  55. Baugh, E.H.; Ke, H.; Levine, A.J.; Bonneau, R.A.; Chan, C.S. Why Are There Hotspot Mutations in the TP53 Gene in Human Cancers? Cell Death Differ. 2018, 25, 154–160. [Google Scholar] [CrossRef]
  56. Levine, A.J. P53, the Cellular Gatekeeper for Growth and Division. Cell 1997, 88, 323–331. [Google Scholar] [CrossRef]
  57. Pacifico, A.; Leone, G. Role of P53 and CDKN2A Inactivation in Human Squamous Cell Carcinomas. J. Biomed. Biotechnol. 2007, 2007, 43418. [Google Scholar] [CrossRef]
  58. Rivlin, N.; Brosh, R.; Oren, M.; Rotter, V. Mutations in the P53 Tumor Suppressor Gene. Genes. Cancer 2011, 2, 466–474. [Google Scholar] [CrossRef] [PubMed]
  59. Roewert-Huber, J.; Stockfleth, E.; Kerl, H. Pathology and Pathobiology of Actinic (Solar) Keratosis—An Update. Br. J. Dermatol. 2007, 157, 18–20. [Google Scholar] [CrossRef] [PubMed]
  60. Hedberg, M.L.; Berry, C.T.; Moshiri, A.S.; Xiang, Y.; Yeh, C.J.; Attilasoy, C.; Capell, B.C.; Seykora, J.T. Molecular Mechanisms of Cutaneous Squamous Cell Carcinoma. Int. J. Mol. Sci. 2022, 23, 3478. [Google Scholar] [CrossRef]
  61. Lefort, K.; Mandinova, A.; Ostano, P.; Kolev, V.; Calpini, V.; Kolfschoten, I.; Devgan, V.; Lieb, J.; Raffoul, W.; Hohl, D.; et al. Notch1 Is a P53 Target Gene Involved in Human Keratinocyte Tumor Suppression through Negative Regulation of ROCK1/2 and MRCKalpha Kinases. Genes Dev. 2007, 21, 562–577. [Google Scholar] [CrossRef]
  62. South, A.P.; Purdie, K.J.; Watt, S.A.; Haldenby, S.; den Breems, N.; Dimon, M.; Arron, S.T.; Kluk, M.J.; Aster, J.C.; McHugh, A.; et al. NOTCH1 Mutations Occur Early during Cutaneous Squamous Cell Carcinogenesis. J. Investig. Dermatol. 2014, 134, 2630–2638. [Google Scholar] [CrossRef]
  63. Boulay, J.-L.; Miserez, A.R.; Zweifel, C.; Sivasankaran, B.; Kana, V.; Ghaffari, A.; Luyken, C.; Sabel, M.; Zerrouqi, A.; Wasner, M.; et al. Loss of NOTCH2 Positively Predicts Survival in Subgroups of Human Glial Brain Tumors. PLoS ONE 2007, 2, e576. [Google Scholar] [CrossRef]
  64. Chu, D.; Zhang, Z.; Zhou, Y.; Wang, W.; Li, Y.; Zhang, H.; Dong, G.; Zhao, Q.; Ji, G. Notch1 and Notch2 Have Opposite Prognostic Effects on Patients with Colorectal Cancer. Ann. Oncol. 2011, 22, 2440–2447. [Google Scholar] [CrossRef] [PubMed]
  65. Fan, X.; Mikolaenko, I.; Elhassan, I.; Ni, X.; Wang, Y.; Ball, D.; Brat, D.J.; Perry, A.; Eberhart, C.G. Notch1 and Notch2 Have Opposite Effects on Embryonal Brain Tumor Growth. Cancer Res. 2004, 64, 7787–7793. [Google Scholar] [CrossRef] [PubMed]
  66. Liu, Z.; Brunskill, E.; Varnum-Finney, B.; Zhang, C.; Zhang, A.; Jay, P.Y.; Bernstein, I.; Morimoto, M.; Kopan, R. The Intracellular Domains of Notch1 and Notch2 Are Functionally Equivalent during Development and Carcinogenesis. Development 2015, 142, 2452–2463. [Google Scholar] [CrossRef]
  67. Danishevich, A.; Bilyalov, A.; Nikolaev, S.; Khalikov, N.; Isaeva, D.; Levina, Y.; Makarova, M.; Nemtsova, M.; Chernevskiy, D.; Sagaydak, O.; et al. CDKN2A Gene Mutations: Implications for Hereditary Cancer Syndromes. Biomedicines 2023, 11, 3343. [Google Scholar] [CrossRef] [PubMed]
  68. Küsters-Vandevelde, H.V.N.; Van Leeuwen, A.; Verdijk, M.A.J.; de Koning, M.N.C.; Quint, W.G.V.; Melchers, W.J.G.; Ligtenberg, M.J.L.; Blokx, W.A.M. CDKN2A but Not TP53 Mutations nor HPV Presence Predict Poor Outcome in Metastatic Squamous Cell Carcinoma of the Skin. Int. J. Cancer 2010, 126, 2123–2132. [Google Scholar] [CrossRef]
  69. PubChem CDKN2A—Cyclin Dependent Kinase Inhibitor 2A (Human). Available online: https://pubchem.ncbi.nlm.nih.gov/gene/CDKN2A/human (accessed on 30 January 2024).
  70. Li, Z.; Gonzalez, C.L.; Wang, B.; Zhang, Y.; Mejia, O.; Katsonis, P.; Lichtarge, O.; Myers, J.N.; El-Naggar, A.K.; Caulin, C. Cdkn2a Suppresses Metastasis in Squamous Cell Carcinomas Induced by the Gain-of-Function Mutant p53R172H. J. Pathol. 2016, 240, 224–234. [Google Scholar] [CrossRef]
  71. Courtney, K.D.; Corcoran, R.B.; Engelman, J.A. The PI3K Pathway As Drug Target in Human Cancer. J. Clin. Oncol. 2010, 28, 1075–1083. [Google Scholar] [CrossRef]
  72. O’Shaughnessy, R.F.L.; Welti, J.C.; Cooke, J.C.; Avilion, A.A.; Monks, B.; Birnbaum, M.J.; Byrne, C. AKT-Dependent HspB1 (Hsp27) Activity in Epidermal Differentiation. J. Biol. Chem. 2007, 282, 17297–17305. [Google Scholar] [CrossRef]
  73. Pankow, S.; Bamberger, C.; Klippel, A.; Werner, S. Regulation of Epidermal Homeostasis and Repair by Phosphoinositide 3-Kinase. J. Cell Sci. 2006, 119, 4033–4046. [Google Scholar] [CrossRef] [PubMed]
  74. Zou, Z.; Tao, T.; Li, H.; Zhu, X. mTOR Signaling Pathway and mTOR Inhibitors in Cancer: Progress and Challenges. Cell Biosci. 2020, 10, 31. [Google Scholar] [CrossRef]
  75. Assad, D.X.; Elias, S.T.; Melo, A.C.; Ferreira, C.G.; De Luca Canto, G.; Guerra, E.N.S. Potential Impact of mTOR Inhibitors on Cervical Squamous Cell Carcinoma: A Systematic Review. Oncol. Lett. 2016, 12, 4107–4116. [Google Scholar] [CrossRef]
  76. Degirmenci, U.; Wang, M.; Hu, J. Targeting Aberrant RAS/RAF/MEK/ERK Signaling for Cancer Therapy. Cells 2020, 9, 198. [Google Scholar] [CrossRef]
  77. McFall, T.; Stites, E.C. Identification of RAS Mutant Biomarkers for EGFR Inhibitor Sensitivity Using a Systems Biochemical Approach. Cell Rep. 2021, 37, 110096. [Google Scholar] [CrossRef] [PubMed]
  78. Li, L.; Li, M.; Xu, S.; Bu, W.; Zhang, M.; Gu, H.; Chen, X. Is Ras a Potential Target in Treatment against Cutaneous Squamous Cell Carcinoma? J. Cancer 2018, 9, 3373–3381. [Google Scholar] [CrossRef]
  79. El-Abaseri, T.B.; Putta, S.; Hansen, L.A. Ultraviolet Irradiation Induces Keratinocyte Proliferation and Epidermal Hyperplasia through the Activation of the Epidermal Growth Factor Receptor. Carcinogenesis 2006, 27, 225–231. [Google Scholar] [CrossRef] [PubMed]
  80. Cañueto, J.; Cardeñoso, E.; García, J.L.; Santos-Briz, Á.; Castellanos-Martín, A.; Fernández-López, E.; Blanco Gómez, A.; Pérez-Losada, J.; Román-Curto, C. Epidermal Growth Factor Receptor Expression Is Associated with Poor Outcome in Cutaneous Squamous Cell Carcinoma. Br. J. Dermatol. 2017, 176, 1279–1287. [Google Scholar] [CrossRef]
  81. Shimizu, T.; Izumi, H.; Oga, A.; Furumoto, H.; Murakami, T.; Ofuji, R.; Muto, M.; Sasaki, K. Epidermal Growth Factor Receptor Overexpression and Genetic Aberrations in Metastatic Squamous-Cell Carcinoma of the Skin. Dermatology 2001, 202, 203–206. [Google Scholar] [CrossRef]
  82. Alter, M.; Satzger, I.; Mattern, A.; Kapp, A.; Gutzmer, R. Treatment of Advanced Cutaneous Squamous Cell Carcinomas with Epidermal Growth Factor Receptor Inhibitors. Dermatology 2013, 227, 289–294. [Google Scholar] [CrossRef]
  83. Maubec, E.; Boubaya, M.; Petrow, P.; Beylot-Barry, M.; Basset-Seguin, N.; Deschamps, L.; Grob, J.-J.; Dréno, B.; Scheer-Senyarich, I.; Bloch-Queyrat, C.; et al. Phase II Study of Pembrolizumab as First-Line, Single-Drug Therapy for Patients with Unresectable Cutaneous Squamous Cell Carcinomas. J. Clin. Oncol. 2020, 38, 3051–3061. [Google Scholar] [CrossRef]
  84. Chen, Z.G.; Saba, N.F.; Teng, Y. The Diverse Functions of FAT1 in Cancer Progression: Good, Bad, or Ugly? J. Exp. Clin. Cancer Res. 2022, 41, 248. [Google Scholar] [CrossRef]
  85. Alonso-Juarranz, M.; Mascaraque, M.; Carrasco, E.; Gracia-Cazaña, T.; De La Sen, O.; Gilaberte, Y.; Gonzalez, S.; Juarranz, Á.; Falahat, F. The Distinctive Features behind the Aggressiveness of Oral and Cutaneous Squamous Cell Carcinomas. Cancers 2023, 15, 3227. [Google Scholar] [CrossRef]
  86. Pastushenko, I.; Mauri, F.; Song, Y.; de Cock, F.; Meeusen, B.; Swedlund, B.; Impens, F.; Van Haver, D.; Opitz, M.; Thery, M.; et al. Fat1 Deletion Promotes Hybrid EMT State, Tumour Stemness and Metastasis. Nature 2021, 589, 448–455. [Google Scholar] [CrossRef] [PubMed]
  87. Hu, X.; Zhai, Y.; Kong, P.; Cui, H.; Yan, T.; Yang, J.; Qian, Y.; Ma, Y.; Wang, F.; Li, H.; et al. FAT1 Prevents Epithelial Mesenchymal Transition (EMT) via MAPK/ERK Signaling Pathway in Esophageal Squamous Cell Cancer. Cancer Lett. 2017, 397, 83–93. [Google Scholar] [CrossRef]
  88. Huang, Z.-L.; Zhang, P.-B.; Zhang, J.-T.; Li, F.; Li, T.-T.; Huang, X.-Y. Comprehensive Genomic Profiling Identifies FAT1 as a Negative Regulator of EMT, CTCs, and Metastasis of Hepatocellular Carcinoma. J. Hepatocell. Carcinoma 2023, 10, 369–382. [Google Scholar] [CrossRef] [PubMed]
  89. Lazar, A.D.; Dinescu, S.; Costache, M. Deciphering the Molecular Landscape of Cutaneous Squamous Cell Carcinoma for Better Diagnosis and Treatment. J. Clin. Med. 2020, 9, 2228. [Google Scholar] [CrossRef]
  90. Liu, H.; Liu, Q.; Ge, Y.; Zhao, Q.; Zheng, X.; Zhao, Y. hTERT Promotes Cell Adhesion and Migration Independent of Telomerase Activity. Sci. Rep. 2016, 6, 22886. [Google Scholar] [CrossRef]
  91. Scott, G.A.; Laughlin, T.S.; Rothberg, P.G. Mutations of the TERT Promoter Are Common in Basal Cell Carcinoma and Squamous Cell Carcinoma. Mod. Pathol. 2014, 27, 516–523. [Google Scholar] [CrossRef]
  92. Yamada-Hishida, H.; Nobeyama, Y.; Nakagawa, H. Correlation of Telomere Length to Malignancy Potential in Non-Melanoma Skin Cancers. Oncol. Lett. 2018, 15, 393–399. [Google Scholar] [CrossRef]
  93. Cheng, K.A.; Kurtis, B.; Babayeva, S.; Zhuge, J.; Tantchou, I.; Cai, D.; Lafaro, R.J.; Fallon, J.T.; Zhong, M. Heterogeneity of TERT Promoter Mutations Status in Squamous Cell Carcinomas of Different Anatomical Sites. Ann. Diagn. Pathol. 2015, 19, 146–148. [Google Scholar] [CrossRef]
  94. Das Mahapatra, K.; Pasquali, L.; Søndergaard, J.N.; Lapins, J.; Nemeth, I.B.; Baltás, E.; Kemény, L.; Homey, B.; Moldovan, L.-I.; Kjems, J.; et al. A Comprehensive Analysis of Coding and Non-Coding Transcriptomic Changes in Cutaneous Squamous Cell Carcinoma. Sci. Rep. 2020, 10, 3637. [Google Scholar] [CrossRef]
  95. Gong, Z.-H.; Zhou, F.; Shi, C.; Xiang, T.; Zhou, C.-K.; Wang, Q.-Q.; Jiang, Y.-S.; Gao, S.-F. miRNA-221 Promotes Cutaneous Squamous Cell Carcinoma Progression by Targeting PTEN. Cell. Mol. Biol. Lett. 2019, 24, 9. [Google Scholar] [CrossRef]
  96. Konicke, K.; López-Luna, A.; Muñoz-Carrillo, J.L.; Servín-González, L.S.; Flores-de la Torre, A.; Olasz, E.; Lazarova, Z. The microRNA Landscape of Cutaneous Squamous Cell Carcinoma. Drug Discov. Today 2018, 23, 864–870. [Google Scholar] [CrossRef] [PubMed]
  97. Li, X.; Huang, K.; Yu, J. Inhibition of microRNA-21 Upregulates the Expression of Programmed Cell Death 4 and Phosphatase Tensin Homologue in the A431 Squamous Cell Carcinoma Cell Line. Oncol. Lett. 2014, 8, 203–207. [Google Scholar] [CrossRef] [PubMed]
  98. Yamane, K.; Jinnin, M.; Etoh, T.; Kobayashi, Y.; Shimozono, N.; Fukushima, S.; Masuguchi, S.; Maruo, K.; Inoue, Y.; Ishihara, T.; et al. Down-Regulation of miR-124/-214 in Cutaneous Squamous Cell Carcinoma Mediates Abnormal Cell Proliferation via the Induction of ERK. J. Mol. Med. 2013, 91, 69–81. [Google Scholar] [CrossRef]
  99. Roskoski, R. Targeting ERK1/2 Protein-Serine/Threonine Kinases in Human Cancers. Pharmacol. Res. 2019, 142, 151–168. [Google Scholar] [CrossRef]
  100. Neu, J.; Dziunycz, P.J.; Dzung, A.; Lefort, K.; Falke, M.; Denzler, R.; Freiberger, S.N.; Iotzova-Weiss, G.; Kuzmanov, A.; Levesque, M.P.; et al. miR-181a Decelerates Proliferation in Cutaneous Squamous Cell Carcinoma by Targeting the Proto-Oncogene KRAS. PLoS ONE 2017, 12, e0185028. [Google Scholar] [CrossRef]
  101. García-Sancha, N.; Corchado-Cobos, R.; Pérez-Losada, J.; Cañueto, J. MicroRNA Dysregulation in Cutaneous Squamous Cell Carcinoma. Int. J. Mol. Sci. 2019, 20, 2181. [Google Scholar] [CrossRef]
  102. Bai, X.; Zhou, Y.; Chen, P.; Yang, M.; Xu, J. MicroRNA-142-5p Induces Cancer Stem Cell-like Properties of Cutaneous Squamous Cell Carcinoma via Inhibiting PTEN. J. Cell Biochem. 2018, 119, 2179–2188. [Google Scholar] [CrossRef] [PubMed]
  103. Lee, C.S.; Bhaduri, A.; Mah, A.; Johnson, W.L.; Ungewickell, A.; Aros, C.J.; Nguyen, C.B.; Rios, E.J.; Siprashvili, Z.; Straight, A.; et al. Recurrent Point Mutations in the Kinetochore Gene KNSTRN in Cutaneous Squamous Cell Carcinoma. Nat. Genet. 2014, 46, 1060–1062. [Google Scholar] [CrossRef] [PubMed]
  104. Schmitz, L.; Grinblat, B.; Novak, B.; Hoeh, A.-K.; Händschke, K.; von Dobbeler, C.; Bierhoff, E.; Szeimies, R.-M.; Gambichler, T.; Torezan, L.; et al. Somatic Mutations in Kinetochore Gene KNSTRN Are Associated with Basal Proliferating Actinic Keratoses and Cutaneous Squamous Cell Carcinoma. J. Eur. Acad. Dermatol. Venereol. 2019, 33, 1535–1540. [Google Scholar] [CrossRef] [PubMed]
  105. Cremolini, C.; Di Bartolomeo, M.; Amatu, A.; Antoniotti, C.; Moretto, R.; Berenato, R.; Perrone, F.; Tamborini, E.; Aprile, G.; Lonardi, S.; et al. BRAF Codons 594 and 596 Mutations Identify a New Molecular Subtype of Metastatic Colorectal Cancer at Favorable Prognosis. Ann. Oncol. 2015, 26, 2092–2097. [Google Scholar] [CrossRef] [PubMed]
  106. Wu, X.; Yan, J.; Dai, J.; Ma, M.; Tang, H.; Yu, J.; Xu, T.; Yu, H.; Si, L.; Chi, Z.; et al. Mutations in BRAF Codons 594 and 596 Predict Good Prognosis in Melanoma. Oncol. Lett. 2017, 14, 3601–3605. [Google Scholar] [CrossRef] [PubMed]
  107. Schleicher, K.; Schramek, D. AJUBA: A Regulator of Epidermal Homeostasis and Cancer. Exp. Dermatol. 2021, 30, 546–559. [Google Scholar] [CrossRef] [PubMed]
  108. ARID2 AT-Rich Interaction Domain 2 [Homo Sapiens (Human)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/196528 (accessed on 5 April 2024).
  109. Stucci, L.S.; Internò, V.; Tucci, M.; Perrone, M.; Mannavola, F.; Palmirotta, R.; Porta, C. The ATM Gene in Breast Cancer: Its Relevance in Clinical Practice. Genes 2021, 12, 727. [Google Scholar] [CrossRef] [PubMed]
  110. Hussain, M.R.M.; Baig, M.; Mohamoud, H.S.A.; Ulhaq, Z.; Hoessli, D.C.; Khogeer, G.S.; Al-Sayed, R.R.; Al-Aama, J.Y. BRAF Gene: From Human Cancers to Developmental Syndromes. Saudi J. Biol. Sci. 2015, 22, 359–373. [Google Scholar] [CrossRef] [PubMed]
  111. Andreassen, P.R.; Seo, J.; Wiek, C.; Hanenberg, H. Understanding BRCA2 Function as a Tumor Suppressor Based on Domain-Specific Activities in DNA Damage Responses. Genes 2021, 12, 1034. [Google Scholar] [CrossRef] [PubMed]
  112. Salvesen, G.S.; Walsh, C.M. Functions of Caspase 8: The Identified and the Mysterious. Semin. Immunol. 2014, 26, 246–252. [Google Scholar] [CrossRef]
  113. Zhu, Y.; Wang, Z.; Li, Y.; Peng, H.; Liu, J.; Zhang, J.; Xiao, X. The Role of CREBBP/EP300 and Its Therapeutic Implications in Hematological Malignancies. Cancers 2023, 15, 1219. [Google Scholar] [CrossRef]
  114. Erbb4 Erb-B2 Receptor Tyrosine Kinase 4 [Mus Musculus (House Mouse)]-Gene-NCBI. Available online: https://www.ncbi.nlm.nih.gov/gene/13869 (accessed on 5 April 2024).
  115. Huang, J.; Gou, H.; Yao, J.; Yi, K.; Jin, Z.; Matsuoka, M.; Zhao, T. The Noncanonical Role of EZH2 in Cancer. Cancer Sci. 2021, 112, 1376. [Google Scholar] [CrossRef] [PubMed]
  116. Ascione, C.M.; Napolitano, F.; Esposito, D.; Servetto, A.; Belli, S.; Santaniello, A.; Scagliarini, S.; Crocetto, F.; Bianco, R.; Formisano, L. Role of FGFR3 in Bladder Cancer: Treatment Landscape and Future Challenges. Cancer Treat. Rev. 2023, 115, 102530. [Google Scholar] [CrossRef]
  117. Hu, X.; Biswas, A.; De, S. KMT2C-Deficient Tumors Have Elevated APOBEC Mutagenesis and Genomic Instability in Multiple Cancers. NAR Cancer 2022, 4, zcac023. [Google Scholar] [CrossRef]
  118. Li, T.; Wang, H.; Xu, J.; Li, C.; Zhang, Y.; Wang, G.; Liu, Y.; Cai, S.; Fang, W.; Li, J.; et al. TGFBR2 Mutation Predicts Resistance to Immune Checkpoint Inhibitors in Patients with Non-Small Cell Lung Cancer. Ther. Adv. Med. Oncol. 2021, 13, 17588359211038477. [Google Scholar] [CrossRef]
  119. Massagué, J. TGFβ Signalling in Context. Nat. Rev. Mol. Cell Biol. 2012, 13, 616–630. [Google Scholar] [CrossRef] [PubMed]
  120. Loesch, R.; Chenane, L.; Colnot, S. ARID2 Chromatin Remodeler in Hepatocellular Carcinoma. Cells 2020, 9, 2152. [Google Scholar] [CrossRef]
  121. Zhang, W.; Liao, Y.; Liu, C.; Liu, L.; Zhou, X. KNSTRN, a Poor Prognostic Biomarker, Affects the Tumor Immune Microenvironment and Immunotherapy Outcomes in Pan-Cancer. Dis. Markers 2023, 2023, 6729717. [Google Scholar] [CrossRef]
  122. Frazzette, N.; Khodadadi-Jamayran, A.; Doudican, N.; Santana, A.; Felsen, D.; Pavlick, A.C.; Tsirigos, A.; Carucci, J.A. Decreased Cytotoxic T Cells and TCR Clonality in Organ Transplant Recipients with Squamous Cell Carcinoma. NPJ Precis. Oncol. 2020, 4, 13. [Google Scholar] [CrossRef]
  123. Kosmidis, M.; Dziunycz, P.; Suárez-Fariñas, M.; Mühleisen, B.; Schärer, L.; Läuchli, S.; Hafner, J.; French, L.E.; Schmidt-Weber, C.; Carucci, J.A.; et al. Immunosuppression Affects CD4+ mRNA Expression and Induces Th2 Dominance in the Microenvironment of Cutaneous Squamous Cell Carcinoma in Organ Transplant Recipients. J. Immunother. 2010, 33, 538–546. [Google Scholar] [CrossRef] [PubMed]
  124. Strobel, S.B.; Safferling, K.; Lahrmann, B.; Hoffmann, J.H.; Enk, A.H.; Hadaschik, E.N.; Grabe, N.; Lonsdorf, A.S. Altered Density, Composition and Microanatomical Distribution of Infiltrating Immune Cells in Cutaneous Squamous Cell Carcinoma of Organ Transplant Recipients. Br. J. Dermatol. 2018, 179, 405–412. [Google Scholar] [CrossRef] [PubMed]
  125. Zilberg, C.; Lyons, J.G.; Gupta, R.; Ferguson, A.; Damian, D.L. The Tumor Immune Microenvironment in Cutaneous Squamous Cell Carcinoma Arising in Organ Transplant Recipients. Ann. Dermatol. 2023, 35, 91–99. [Google Scholar] [CrossRef] [PubMed]
  126. Rashtak, S.; Dierkhising, R.A.; Kremers, W.K.; Peters, S.G.; Cassivi, S.D.; Otley, C.C. Incidence and Risk Factors for Skin Cancer Following Lung Transplantation. J. Am. Acad. Dermatol. 2015, 72, 92–98. [Google Scholar] [CrossRef] [PubMed]
  127. Friman, T.K.; Jäämaa-Holmberg, S.; Åberg, F.; Helanterä, I.; Halme, M.; Pentikäinen, M.O.; Nordin, A.; Lemström, K.B.; Jahnukainen, T.; Räty, R.; et al. Cancer Risk and Mortality after Solid Organ Transplantation: A Population-Based 30-Year Cohort Study in Finland. Int. J. Cancer 2022, 150, 1779–1791. [Google Scholar] [CrossRef] [PubMed]
  128. Anderson, N.M.; Simon, M.C. The Tumor Microenvironment. Curr. Biol. 2020, 30, R921–R925. [Google Scholar] [CrossRef] [PubMed]
  129. Martínez-Nieto, G.A.; Teppo, H.-R.; Petrelius, N.; Izzi, V.; Devarajan, R.; Petäistö, T.; Liu, H.; Kim, K.S.; Karppinen, S.-M.; Ruotsalainen, H.; et al. Upregulated Integrin A11 in the Stroma of Cutaneous Squamous Cell Carcinoma Promotes Skin Carcinogenesis. Front. Oncol. 2022, 12, 981009. [Google Scholar] [CrossRef]
  130. Frantz, C.; Stewart, K.M.; Weaver, V.M. The Extracellular Matrix at a Glance. J. Cell Sci. 2010, 123, 4195–4200. [Google Scholar] [CrossRef] [PubMed]
  131. Kular, J.K.; Basu, S.; Sharma, R.I. The Extracellular Matrix: Structure, Composition, Age-Related Differences, Tools for Analysis and Applications for Tissue Engineering. J. Tissue Eng. 2014, 5, 2041731414557112. [Google Scholar] [CrossRef]
  132. Peake, N.J.; Khawaja, K.; Myers, A.; Jones, D.; Cawston, T.E.; Rowan, A.D.; Foster, H.E. Levels of Matrix Metalloproteinase (MMP)-1 in Paired Sera and Synovial Fluids of Juvenile Idiopathic Arthritis Patients: Relationship to Inflammatory Activity, MMP-3 and Tissue Inhibitor of Metalloproteinases-1 in a Longitudinal Study. Rheumatology 2005, 44, 1383–1389. [Google Scholar] [CrossRef]
  133. Wang, H.; Li, H.; Yan, Q.; Gao, S.; Gao, J.; Wang, Z.; Sun, Y. Serum Matrix Metalloproteinase-13 as a Diagnostic Biomarker for Cutaneous Squamous Cell Carcinoma. BMC Cancer 2021, 21, 816. [Google Scholar] [CrossRef]
  134. Fromme, J.E.; Zigrino, P. The Role of Extracellular Matrix Remodeling in Skin Tumor Progression and Therapeutic Resistance. Front. Mol. Biosci. 2022, 9, 864302. [Google Scholar] [CrossRef]
  135. Lu, P.; Takai, K.; Weaver, V.M.; Werb, Z. Extracellular Matrix Degradation and Remodeling in Development and Disease. Cold Spring Harb. Perspect. Biol. 2011, 3, a005058. [Google Scholar] [CrossRef]
  136. Georgescu, S.R.; Tampa, M.; Mitran, C.I.; Mitran, M.I.; Caruntu, C.; Caruntu, A.; Lupu, M.; Matei, C.; Constantin, C.; Neagu, M. Tumour Microenvironment in Skin Carcinogenesis. In Tumor Microenvironments in Organs: From the Brain to the Skin—Part A; Birbrair, A., Ed.; Springer International Publishing: Cham, Switzerland, 2020; pp. 123–142. ISBN 978-3-030-36214-0. [Google Scholar]
  137. Kambayashi, Y.; Fujimura, T.; Aiba, S. Comparison of Immunosuppressive and Immunomodulatory Cells in Keratoacanthoma and Cutaneous Squamous Cell Carcinoma. Acta Derm.-Venereol. 2013, 93, 663–668. [Google Scholar] [CrossRef] [PubMed]
  138. Pettersen, J.S.; Fuentes-Duculan, J.; Suárez-Fariñas, M.; Pierson, K.C.; Pitts-Kiefer, A.; Fan, L.; Belkin, D.A.; Wang, C.Q.F.; Bhuvanendran, S.; Johnson-Huang, L.M.; et al. Tumor-Associated Macrophages in the Cutaneous SCC Microenvironment Are Heterogeneously Activated. J. Investig. Dermatol. 2011, 131, 1322–1330. [Google Scholar] [CrossRef]
  139. Ahmed Haji Omar, A.; Haglund, C.; Virolainen, S.; Häyry, V.; Atula, T.; Kontio, R.; Salo, T.; Sorsa, T.; Hagström, J. MMP-7, MMP-8, and MMP-9 in Oral and Cutaneous Squamous Cell Carcinomas. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2015, 119, 459–467. [Google Scholar] [CrossRef]
  140. Ricard-Blum, S. The Collagen Family. Cold Spring Harb. Perspect. Biol. 2011, 3, a004978. [Google Scholar] [CrossRef]
  141. Karppinen, S.-M.; Honkanen, H.-K.; Heljasvaara, R.; Riihilä, P.; Autio-Harmainen, H.; Sormunen, R.; Harjunen, V.; Väisänen, M.-R.; Väisänen, T.; Hurskainen, T.; et al. Collagens XV and XVIII Show Different Expression and Localisation in Cutaneous Squamous Cell Carcinoma: Type XV Appears in Tumor Stroma, While XVIII Becomes Upregulated in Tumor Cells and Lost from Microvessels. Exp. Dermatol. 2016, 25, 348–354. [Google Scholar] [CrossRef]
  142. Martins, V.L.; Caley, M.P.; Moore, K.; Szentpetery, Z.; Marsh, S.T.; Murrell, D.F.; Kim, M.H.; Avari, M.; McGrath, J.A.; Cerio, R.; et al. Suppression of TGFβ and Angiogenesis by Type VII Collagen in Cutaneous SCC. J. Natl. Cancer Inst. 2016, 108, djv293. [Google Scholar] [CrossRef]
  143. Bhowmick, N.A.; Neilson, E.G.; Moses, H.L. Stromal Fibroblasts in Cancer Initiation and Progression. Nature 2004, 432, 332–337. [Google Scholar] [CrossRef]
  144. Gallego-Rentero, M.; Gutiérrez-Pérez, M.; Fernández-Guarino, M.; Mascaraque, M.; Portillo-Esnaola, M.; Gilaberte, Y.; Carrasco, E.; Juarranz, Á. TGFβ1 Secreted by Cancer-Associated Fibroblasts as an Inductor of Resistance to Photodynamic Therapy in Squamous Cell Carcinoma Cells. Cancers 2021, 13, 5613. [Google Scholar] [CrossRef]
  145. Turley, S.J.; Cremasco, V.; Astarita, J.L. Immunological Hallmarks of Stromal Cells in the Tumour Microenvironment. Nat. Rev. Immunol. 2015, 15, 669–682. [Google Scholar] [CrossRef] [PubMed]
  146. Hirayama, D.; Iida, T.; Nakase, H. The Phagocytic Function of Macrophage-Enforcing Innate Immunity and Tissue Homeostasis. Int. J. Mol. Sci. 2017, 19, 92. [Google Scholar] [CrossRef]
  147. Wang, Y.-C.; He, F.; Feng, F.; Liu, X.-W.; Dong, G.-Y.; Qin, H.-Y.; Hu, X.-B.; Zheng, M.-H.; Liang, L.; Feng, L.; et al. Notch Signaling Determines the M1 versus M2 Polarization of Macrophages in Antitumor Immune Responses. Cancer Res. 2010, 70, 4840–4849. [Google Scholar] [CrossRef] [PubMed]
  148. Linde, N.; Lederle, W.; Depner, S.; van Rooijen, N.; Gutschalk, C.M.; Mueller, M.M. Vascular Endothelial Growth Factor-Induced Skin Carcinogenesis Depends on Recruitment and Alternative Activation of Macrophages. J. Pathol. 2012, 227, 17–28. [Google Scholar] [CrossRef]
  149. Moussai, D.; Mitsui, H.; Pettersen, J.S.; Pierson, K.C.; Shah, K.R.; Suárez-Fariñas, M.; Cardinale, I.R.; Bluth, M.J.; Krueger, J.G.; Carucci, J.A. The Human Cutaneous Squamous Cell Carcinoma Microenvironment Is Characterized by Increased Lymphatic Density and Enhanced Expression of Macrophage-Derived VEGF-C. J. Investig. Dermatol. 2011, 131, 229–236. [Google Scholar] [CrossRef] [PubMed]
  150. Mantovani, A.; Sozzani, S.; Locati, M.; Allavena, P.; Sica, A. Macrophage Polarization: Tumor-Associated Macrophages as a Paradigm for Polarized M2 Mononuclear Phagocytes. Trends Immunol. 2002, 23, 549–555. [Google Scholar] [CrossRef] [PubMed]
  151. Cyrus, N.; Mai-Anh Bui, C.; Yao, X.; Kohn, L.L.; Galan, A.; Rhebergen, A.M.; Colegio, O.R. Density and Polarization States of Tumor-Associated Macrophages in Human Cutaneous Squamous Cell Carcinomas Arising in Solid Organ Transplant Recipients. Dermatol. Surg. 2016, 42 (Suppl. S1), S18–S23. [Google Scholar] [CrossRef]
  152. Boutilier, A.J.; Elsawa, S.F. Macrophage Polarization States in the Tumor Microenvironment. Int. J. Mol. Sci. 2021, 22, 6995. [Google Scholar] [CrossRef]
  153. El-Rouby, D.H. Association of Macrophages with Angiogenesis in Oral Verrucous and Squamous Cell Carcinomas. J. Oral Pathol. Med. 2010, 39, 559–564. [Google Scholar] [CrossRef]
  154. Leek, R.D.; Lewis, C.E.; Whitehouse, R.; Greenall, M.; Clarke, J.; Harris, A.L. Association of Macrophage Infiltration with Angiogenesis and Prognosis in Invasive Breast Carcinoma. Cancer Res. 1996, 56, 4625–4629. [Google Scholar] [PubMed]
  155. Lissbrant, I.F.; Stattin, P.; Wikstrom, P.; Damber, J.E.; Egevad, L.; Bergh, A. Tumor Associated Macrophages in Human Prostate Cancer: Relation to Clinicopathological Variables and Survival. Int. J. Oncol. 2000, 17, 445–451. [Google Scholar] [CrossRef]
  156. Saeidi, V.; Doudican, N.; Carucci, J.A. Understanding the Squamous Cell Carcinoma Immune Microenvironment. Front. Immunol. 2023, 14, 1084873. [Google Scholar] [CrossRef]
  157. Costa, N.L.; Valadares, M.C.; Souza, P.P.C.; Mendonça, E.F.; Oliveira, J.C.; Silva, T.A.; Batista, A.C. Tumor-Associated Macrophages and the Profile of Inflammatory Cytokines in Oral Squamous Cell Carcinoma. Oral Oncol. 2013, 49, 216–223. [Google Scholar] [CrossRef]
  158. Kumar, A.T.; Knops, A.; Swendseid, B.; Martinez-Outschoom, U.; Harshyne, L.; Philp, N.; Rodeck, U.; Luginbuhl, A.; Cognetti, D.; Johnson, J.; et al. Prognostic Significance of Tumor-Associated Macrophage Content in Head and Neck Squamous Cell Carcinoma: A Meta-Analysis. Front. Oncol. 2019, 9, 656. [Google Scholar] [CrossRef]
  159. Fujimura, T.; Kambayashi, Y.; Fujisawa, Y.; Hidaka, T.; Aiba, S. Tumor-Associated Macrophages: Therapeutic Targets for Skin Cancer. Front. Oncol. 2018, 8, 3. [Google Scholar] [CrossRef]
  160. Zhang, S.; Fujita, H.; Mitsui, H.; Yanofsky, V.R.; Fuentes-Duculan, J.; Pettersen, J.S.; Suárez-Fariñas, M.; Gonzalez, J.; Wang, C.Q.F.; Krueger, J.G.; et al. Increased Tc22 and Treg/CD8 Ratio Contribute to Aggressive Growth of Transplant Associated Squamous Cell Carcinoma. PLoS ONE 2013, 8, e62154. [Google Scholar] [CrossRef]
  161. Curiel, T.J.; Coukos, G.; Zou, L.; Alvarez, X.; Cheng, P.; Mottram, P.; Evdemon-Hogan, M.; Conejo-Garcia, J.R.; Zhang, L.; Burow, M.; et al. Specific Recruitment of Regulatory T Cells in Ovarian Carcinoma Fosters Immune Privilege and Predicts Reduced Survival. Nat. Med. 2004, 10, 942–949. [Google Scholar] [CrossRef] [PubMed]
  162. Bates, G.J.; Fox, S.B.; Han, C.; Leek, R.D.; Garcia, J.F.; Harris, A.L.; Banham, A.H. Quantification of Regulatory T Cells Enables the Identification of High-Risk Breast Cancer Patients and Those at Risk of Late Relapse. J. Clin. Oncol. 2006, 24, 5373–5380. [Google Scholar] [CrossRef] [PubMed]
  163. Fu, J.; Xu, D.; Liu, Z.; Shi, M.; Zhao, P.; Fu, B.; Zhang, Z.; Yang, H.; Zhang, H.; Zhou, C.; et al. Increased Regulatory T Cells Correlate with CD8 T-Cell Impairment and Poor Survival in Hepatocellular Carcinoma Patients. Gastroenterology 2007, 132, 2328–2339. [Google Scholar] [CrossRef]
  164. Gobert, M.; Treilleux, I.; Bendriss-Vermare, N.; Bachelot, T.; Goddard-Leon, S.; Arfi, V.; Biota, C.; Doffin, A.C.; Durand, I.; Olive, D.; et al. Regulatory T Cells Recruited through CCL22/CCR4 Are Selectively Activated in Lymphoid Infiltrates Surrounding Primary Breast Tumors and Lead to an Adverse Clinical Outcome. Cancer Res. 2009, 69, 2000–2009. [Google Scholar] [CrossRef]
  165. Lai, C.; August, S.; Albibas, A.; Behar, R.; Cho, S.-Y.; Polak, M.E.; Theaker, J.; MacLeod, A.S.; French, R.R.; Glennie, M.J.; et al. OX40+ Regulatory T Cells in Cutaneous Squamous Cell Carcinoma Suppress Effector T Cell Responses and Associate with Metastatic Potential. Clin. Cancer Res. 2016, 22, 4236–4248. [Google Scholar] [CrossRef]
  166. Zhang, S.; Wu, M.; Wang, F. Immune Regulation by CD8+ Treg Cells: Novel Possibilities for Anticancer Immunotherapy. Cell Mol. Immunol. 2018, 15, 805–807. [Google Scholar] [CrossRef]
  167. Ji, A.L.; Rubin, A.J.; Thrane, K.; Jiang, S.; Reynolds, D.L.; Meyers, R.M.; Guo, M.G.; George, B.M.; Mollbrink, A.; Bergenstråhle, J.; et al. Multimodal Analysis of Composition and Spatial Architecture in Human Squamous Cell Carcinoma. Cell 2020, 182, 497–514.e22. [Google Scholar] [CrossRef] [PubMed]
  168. Adhikary, G.; Heipertz, E.L.; Preradovic, M.; Chen, X.; Xu, W.; Newland, J.J.; Kaur, N.; Vemuri, M.C.; Eckert, R.L. Natural Killer Cells Suppress Human Cutaneous Squamous Cell Carcinoma Cancer Cell Survival and Tumor Growth. Mol. Carcinog. 2023, 62, 845–854. [Google Scholar] [CrossRef] [PubMed]
  169. Luci, C.; Bihl, F.; Bourdely, P.; Khou, S.; Popa, A.; Meghraoui-Kheddar, A.; Vermeulen, O.; Elaldi, R.; Poissonnet, G.; Sudaka, A.; et al. Cutaneous Squamous Cell Carcinoma Development Is Associated with a Temporal Infiltration of ILC1 and NK Cells with Immune Dysfunctions. J. Investig. Dermatol. 2021, 141, 2369–2379. [Google Scholar] [CrossRef]
  170. de Souza Fernandez Pereira, M.; Carr, D.R.; Gatti-Mays, M.E.; Olsen, M.R.; Setty, B.A.; Shahwan, K.T.; Lee, D.A. Natural Killer Cell Recognition and Control of Epithelial Cancers. Cancer J. 2022, 28, 263–269. [Google Scholar] [CrossRef]
  171. Ilyas, M.; Costello, C.M.; Sharma, A. Exploring the Relationship between Natural Killer Cells and Cutaneous Squamous Cell Carcinoma Development. JAAD Case Rep. 2017, 3, 364–366. [Google Scholar] [CrossRef] [PubMed]
  172. Khou, S.; Popa, A.; Luci, C.; Bihl, F.; Meghraoui-Kheddar, A.; Bourdely, P.; Salavagione, E.; Cosson, E.; Rubod, A.; Cazareth, J.; et al. Tumor-Associated Neutrophils Dampen Adaptive Immunity and Promote Cutaneous Squamous Cell Carcinoma Development. Cancers 2020, 12, 1860. [Google Scholar] [CrossRef]
  173. Seddon, A.; Hock, B.; Miller, A.; Frei, L.; Pearson, J.; McKenzie, J.; Simcock, J.; Currie, M. Cutaneous Squamous Cell Carcinomas with Markers of Increased Metastatic Risk Are Associated with Elevated Numbers of Neutrophils and/or Granulocytic Myeloid Derived Suppressor Cells. J. Dermatol. Sci. 2016, 83, 124–130. [Google Scholar] [CrossRef]
  174. Grob, J.-J.; Gonzalez, R.; Basset-Seguin, N.; Vornicova, O.; Schachter, J.; Joshi, A.; Meyer, N.; Grange, F.; Piulats, J.M.; Bauman, J.R.; et al. Pembrolizumab Monotherapy for Recurrent or Metastatic Cutaneous Squamous Cell Carcinoma: A Single-Arm Phase II Trial (KEYNOTE-629). J. Clin. Oncol. 2020, 38, 2916. [Google Scholar] [CrossRef]
  175. Maubec, E.; Petrow, P.; Scheer-Senyarich, I.; Duvillard, P.; Lacroix, L.; Gelly, J.; Certain, A.; Duval, X.; Crickx, B.; Buffard, V.; et al. Phase II Study of Cetuximab as First-Line Single-Drug Therapy in Patients with Unresectable Squamous Cell Carcinoma of the Skin. J. Clin. Oncol. 2011, 29, 3419–3426. [Google Scholar] [CrossRef]
  176. 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. 2014, 25, 2047–2052. [Google Scholar] [CrossRef]
  177. Gold, K.A.; Kies, M.S.; William, N.; William, J.; 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 II Clinical Trial. Cancer 2018, 124, 2169. [Google Scholar] [CrossRef] [PubMed]
  178. 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] [PubMed]
  179. Acevedo-Henao, C.M.; Valette, G.; Miglierini, P.; Lefur, E.; Pradier, O. Radiotherapy Combined with Cetuximab for Locally Advanced Head and Neck Cancer: Results and Toxicity. Cancer Radiother. 2012, 16, 601–603. [Google Scholar] [CrossRef]
  180. Sacco, A.G.; Chen, R.; Worden, F.P.; Wong, D.J.L.; Adkins, D.; Swiecicki, P.; Chai-Ho, W.; Oppelt, P.; Ghosh, D.; Bykowski, J.; et al. Pembrolizumab plus Cetuximab in Patients with Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma: An Open-Label, Multi-Arm, Non-Randomised, Multicentre, Phase 2 Trial. Lancet Oncol. 2021, 22, 883–892. [Google Scholar] [CrossRef] [PubMed]
  181. Chung, C.H.; Li, J.; Steuer, C.E.; Bhateja, P.; Johnson, M.; Masannat, J.; Poole, M.I.; Song, F.; Hernandez-Prera, J.C.; Molina, H.; et al. Phase II Multi-Institutional Clinical Trial Result of Concurrent Cetuximab and Nivolumab in Recurrent and/or Metastatic Head and Neck Squamous Cell Carcinoma. Clin. Cancer Res. 2022, 28, 2329–2338. [Google Scholar] [CrossRef]
  182. Esfahani, K.; Roudaia, L.; Buhlaiga, N.; Del Rincon, S.V.; Papneja, N.; Miller, W.H. A Review of Cancer Immunotherapy: From the Past, to the Present, to the Future. Curr. Oncol. 2020, 27, S87–S97. [Google Scholar] [CrossRef] [PubMed]
  183. 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]
  184. Patel, R.; Chang, A.L.S. Immune Checkpoint Inhibitors for Treating Advanced Cutaneous Squamous Cell Carcinoma. Am. J. Clin. Dermatol. 2019, 20, 477–482. [Google Scholar] [CrossRef]
  185. Center for Drug Evaluation and Research. FDA Approves Cemiplimab-Rwlc for Metastatic or Locally Advanced Cutaneous Squamous Cell Carcinoma; FDA: Silver Spring, MD, USA, 2019. [Google Scholar]
  186. Patel, S.P.; Othus, M.; Chen, Y.; Wright, G.P.; Yost, K.J.; Hyngstrom, J.R.; Hu-Lieskovan, S.; Lao, C.D.; Fecher, L.A.; Truong, T.-G.; et al. Neoadjuvant–Adjuvant or Adjuvant-Only Pembrolizumab in Advanced Melanoma. N. Engl. J. Med. 2023, 388, 813–823. [Google Scholar] [CrossRef] [PubMed]
  187. Marin-Acevedo, J.A.; Withycombe, B.M.; Kim, Y.; Brohl, A.S.; Eroglu, Z.; Markowitz, J.; Tarhini, A.A.; Tsai, K.Y.; Khushalani, N.I. Cetuximab for Immunotherapy-Refractory/Ineligible Cutaneous Squamous Cell Carcinoma. Cancers 2023, 15, 3180. [Google Scholar] [CrossRef] [PubMed]
  188. Wollina, U. Update of Cetuximab for Non-Melanoma Skin Cancer. Expert Opin. Biol. Ther. 2014, 14, 271–276. [Google Scholar] [CrossRef] [PubMed]
  189. Chang, M.; Samlowski, W.; Meoz, R. Effectiveness and Toxicity of Cetuximab with Concurrent RT in Locally Advanced Cutaneous Squamous Cell Skin Cancer: A Case Series. Oncotarget 2023, 14, 709–718. [Google Scholar] [CrossRef]
  190. Bonner, J.A.; Harari, P.M.; Giralt, J.; Azarnia, N.; Shin, D.M.; Cohen, R.B.; Jones, C.U.; Sur, R.; Raben, D.; Jassem, J.; et al. Radiotherapy plus Cetuximab for Squamous-Cell Carcinoma of the Head and Neck. N. Engl. J. Med. 2006, 354, 567–578. [Google Scholar] [CrossRef]
  191. Hsu, E.; Eton, O.; Patel, A.V.; Cartun, R.; Earle, J.S.; Mnayer, L.O.; Kotowitz, J.; Yu, P.P. Combining Panitumumab with Anti-PD-1 Antibody in Cutaneous Squamous Cell Carcinoma of the Head and Neck after Inadequate Response to Anti-PD-1 Antibody Alone. J. Drugs Dermatol. 2021, 20, 901–904. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Visualization of the pathway and driver gene interactions in cSCC. Figure references [1,5,27,28,29,31,34,35,36,37,38,39,40,41,42,43,44,45,46,47].
Figure 1. Visualization of the pathway and driver gene interactions in cSCC. Figure references [1,5,27,28,29,31,34,35,36,37,38,39,40,41,42,43,44,45,46,47].
Cancers 16 02904 g001
Table 1. Major validated driver genes and pathways in cSCC; Genes and pathways cited in four or more primary papers as mutated or dysregulated in cSCC, and consistently validated as key components in cSCC tumorigenesis. Description references [1,5,27,28,29,31,34,35,36,37,38,39,40,41,42,43,44,45,46,47].
Table 1. Major validated driver genes and pathways in cSCC; Genes and pathways cited in four or more primary papers as mutated or dysregulated in cSCC, and consistently validated as key components in cSCC tumorigenesis. Description references [1,5,27,28,29,31,34,35,36,37,38,39,40,41,42,43,44,45,46,47].
Gene(s)General DescriptionReferences
TP53Tumor protein p53; regulates genome stability, cell proliferation, and apoptosis[1,5,27,31,36,37,40,45]
NOTCH 1,2,3Notch receptor; regulates cell signaling, differentiation, cell cycle arrest, and apoptosis[1,5,27,31,36,37,45]
CDKN2ACyclin-dependent kinase inhibitor 2A;
regulates genomic stability, Cell cycle control, and cell proliferation
[1,5,27,31,40,48]
RAS/RAF/MEK/MAPK(ERK) PathwayRegulates cell growth, migration, angiogenesis, proliferation, differentiation, apoptosis, and metabolism[5,28,31,34,48]
PI3K/AKT/mTOR PathwayRegulates cell proliferation, growth differentiation, and cell apoptosis inhibition[5,28,31,34,37,48]
HRAS; KRASProto-oncogenes, GTPase; regulates cell proliferation, migration, growth, survival, and differentiation[1,5,31,36,37,40,49]; [1,5,28,31,36,37,40,49]
EGFREpidermal growth factor receptor; activates signaling pathways, regulates tissue regeneration, construction, homeostasis, cellular proliferation, apoptosis, and differentiation[31,35,37,50]
FAT1FAT atypical Cadherin 1; regulates mitochondrial respiration, cellular migration, and proliferation[1,5,27,42]
PTENPhosphatase and tensin homolog; activates PI3K/AKT/mTOR pathway and facilitates cellular migration, growth, proliferation, signaling, protein synthesis, and DNA repair[5,27,31,46]
Table 2. Additional genes cited in 2–3 primary sources as mutationally relevant and potential contributors to cSCC tumorigenesis. Description references [107,108,109,110,111,112,113,114,115,116,117,118,119,120,121].
Table 2. Additional genes cited in 2–3 primary sources as mutationally relevant and potential contributors to cSCC tumorigenesis. Description references [107,108,109,110,111,112,113,114,115,116,117,118,119,120,121].
Gene(s) Description References
CCND1Cyclin D1, a regulator of CDK kinases; regulates cell cycle and metastasis [5,31,37]
CREBBPCREB-binding protein; regulates tumor response, cell growth, and division [5,31,42]
EP300E1A-binding protein P300; cell growth, proliferation, maturation, differentiation [5,31,42]
ERBB4Erb-b2 receptor tyrosine kinase 4; tumor suppressor, cellular response to EGFR[31,37,42]
EZH2Enhancer of zeste 2 polycomb repressive complex 2; silences tumor suppressor genes and regulates apoptosis, cell proliferation, and cycle regulation[5,31,42]
FGFR3Fibroblast growth factor receptor 3; regulates cell proliferation, survival, migration, and angiogenesis[31,40,50]
KMT2C/KMT2D (MLL3)Histone lysine methyltransferase 2C/2D; regulates transcriptional coactivation, genome maintenance, and histone methylation[1,37,42]
TGFBR2Transforming growth factor beta receptor 2; regulates cell differentiation, proliferation, maturation, and tissue homeostasis [36,38,47]
AJUBAAjuba LIM protein; regulates epidermal cell differentiation and homeostasis [1,5]
ARID2 AT-rich interaction domain 2; chromatin remodeling[5,31]
ATMAtaxia telangiectasia mutated; regulates stress response and DNA repair [37,42]
BRAFSerine/threonine protein kinase; regulates MAP/ERK pathway, cell division, differentiation, and secretion [31,42]
BRCA2Breast cancer gene 2; regulates genomic stability, DNA repair, and damage response[37,39]
CASP8Cysteine-aspartic acid protease; regulates cell survival and apoptosis [1,5]
KNSTRNKinetochore-localized astrin/SPAG5-binding protein; regulates chromosomal stability and division[103,121]
LRP1BLow-density lipoprotein receptor-related protein 1B; regulates cell signaling and migration [37,39]
NFE2L2Nuclear factor erythroid 2-related transcription factor 2; regulates oxidative stress and inflammatory response [1,5]
CARD11Caspase recruitment domain family, member protein 11; regulates immune response through lymphocyte activation[31,43]
MYCMyelocytomatosis oncogene; regulates mRNA translation, stress response, immune response, cell cycle, proliferation, differentiation, apoptosis[31,40]
RIPK4Receptor-interacting serine/threonine kinase protein 4; regulates keratinocyte differentiation, cutaneous inflammation, and wound repair[1,31]
Table 3. Phase II clinical trials conducted on cSCC or HNSCC with the respective overall response rates (ORRs). ORRs were calculated using the number of patients who showed partial or complete response, using the intent to treat population. * ORRs for the neoadjuvant trial are reported using major pathological responses (<10% tumor volume) and complete pathological responses.
Table 3. Phase II clinical trials conducted on cSCC or HNSCC with the respective overall response rates (ORRs). ORRs were calculated using the number of patients who showed partial or complete response, using the intent to treat population. * ORRs for the neoadjuvant trial are reported using major pathological responses (<10% tumor volume) and complete pathological responses.
Treatment(s)Patient PopulationORRReferences
Immunotherapy Monotherapy: cemiplimab advanced metastatic cSCC 47% (n = 59)[11]
Immunotherapy Monotherapy: pembrolizumabrecurrent or metastatic cSCC 34% (n = 105)[174]
Immunotherapy Monotherapy: pembrolizumabprimary unresectable cSCC 42% (n = 57)[83]
Neoadjuvant Immunotherapy: cemiplimabadvanced resectable cSCC63% (n = 79) *[12]
EGFR Monotherapy: cetuximabprimary unresectable cSCC 28% (n = 36)[175]
EGFR Monotherapy: panitumumabadvanced, metastatic, or
recurrent cSCC
31% (n = 16)[176]
EGFR Monotherapy: erlotinibrecurrent or metastatic cSCC 8% (n = 39)[177]
EGFR Monotherapy: gefitinibrecurrent and or metastatic cSCC 15% (n = 40)[178]
Radiation + EGFR Inhibitors: cetuximablocally advanced HNSCC91% (n = 36)[179]
Immunotherapy + EGFR Inhibitors:
pembrolizumab and cetuximab
recurrent or metastatic HNSCC45% (n = 33)[180]
Immunotherapy + EGFR Inhibitors:
nivolamb and cetuximab
previously treated recurrent or metastatic HNSCC;
first-line recurrent or metastatic HNSCC
23% (n = 47);
33% (n = 48)
[181]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hosseini, T.M.; Park, S.J.; Guo, T. The Mutational and Microenvironmental Landscape of Cutaneous Squamous Cell Carcinoma: A Review. Cancers 2024, 16, 2904. https://doi.org/10.3390/cancers16162904

AMA Style

Hosseini TM, Park SJ, Guo T. The Mutational and Microenvironmental Landscape of Cutaneous Squamous Cell Carcinoma: A Review. Cancers. 2024; 16(16):2904. https://doi.org/10.3390/cancers16162904

Chicago/Turabian Style

Hosseini, Tara M., Soo J. Park, and Theresa Guo. 2024. "The Mutational and Microenvironmental Landscape of Cutaneous Squamous Cell Carcinoma: A Review" Cancers 16, no. 16: 2904. https://doi.org/10.3390/cancers16162904

APA Style

Hosseini, T. M., Park, S. J., & Guo, T. (2024). The Mutational and Microenvironmental Landscape of Cutaneous Squamous Cell Carcinoma: A Review. Cancers, 16(16), 2904. https://doi.org/10.3390/cancers16162904

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