Next Article in Journal
Socioeconomic Status and Glycemic Control in Type 2 Diabetes; Race by Gender Differences
Previous Article in Journal
Senescent Nephropathy: The New Renal Syndrome
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Updates on the Management of Non-Melanoma Skin Cancer (NMSC)

1
Division of Plastic and Reconstructive Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
2
Division of Plastic and Maxillofacial Surgery, Children’s Hospital of Los Angeles, Los Angeles, CA 90027, USA
3
Keck School of Medicine of University of Southern California, Los Angeles, CA 91001, USA
*
Author to whom correspondence should be addressed.
Healthcare 2017, 5(4), 82; https://doi.org/10.3390/healthcare5040082
Submission received: 8 September 2017 / Revised: 20 October 2017 / Accepted: 24 October 2017 / Published: 1 November 2017

Abstract

:
Non-melanoma skin cancers (NMSCs) are the most common malignancy worldwide, of which 99% are basal cell carcinomas (BCCs) and squamous cell carcinomas (SCCs) of skin. NMSCs are generally considered a curable diseases, yet they currently pose an increasing global healthcare problem due to rising incidence. This has led to a shift in emphasis on prevention of NMSCs with development of various skin cancer prevention programs worldwide. This article aims to summarize the most recent changes and advances made in NMSC management with a focus on prevention, screening, diagnosis, and staging.

1. Introduction

Non-melanoma skin cancers (NMSC), also known as keratinocyte cancers, are the most common malignancy worldwide [1,2]. The incidence rate is the highest in Australia with more than 1000 per 100,000 person-years, followed by Europe with 98 per 100,000 person-years [1,2,3,4]. The number of people with NMSC increased from 2.4 million to 3.3 million from 2006 to 2012 in the USA [5]. Basal cell carcinoma (BCC) and squamous cell carcinoma (SCC) of the skin make up 99% of all NMSC, with BCC being 3 to 5 times more common than SCC [1,4,5,6,7]. Other much less common forms of NMSC include: Merkel cell carcinoma (MCC), primary cutaneous B-cell lymphoma, Kaposi sarcoma, carcinosarcoma, and dermatofibrosarcoma [6,8]. Although NMSCs are generally considered to be curable, they pose a vast problem for healthcare worldwide due to rising incidence [1,9,10,11]. Some NMSCs are associated with fatal outcomes. In particular, SCC is responsible for the majority of NMSC-related deaths [9]. The purpose of this paper is to discuss the updates on current management of NMSC and review the latest American Joint Committee on Cancers (AJCC) and National Comprehensive Cancer Network (NCCN) guidelines.

2. Brief Overview of Etiology, Risk Factors and Staging

2.1. Etiology and Risk Factors

The hypothesis that sunlight leads to skin cancer has existed for more than a century, however, the mathematical relationship between ultraviolet (UV) exposure and the risk of skin cancers was first described by Fears et al. in 1977 [12,13]. The authors suggested that while melanoma was primarily caused by intermittent sun exposure, keratinocyte cancers were related to cumulative doses of ultraviolent light [14,15]. Additional risk factors for development of NMSC include radiation therapy, lower Fitzpatrick skin types 1–4, prolonged immunosuppression, human immunodeficiency virus (HIV), human papilloma virus (HPV), and a diagnosis of certain syndromes or genetic disorders [16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34]. Unusual changes in peristomal skin of ileostomy and gastrostomy, burn scars, chronic inflammatory dermatologic conditions, and non-healing ulcers should also raise a suspicion for SCC, particularly Majolin’s ulcers [35,36,37,38,39,40,41,42]. Moreover, it has been well documented that male gender, tumor location in the trunk and extremities, superficial histologic subtype, younger age at first diagnosis of BCC, and red hair phenotype are associated with a higher risk of having multiple lesions [43,44,45,46,47,48].
Immunosuppressed patients have a particularly increased risk of developing NMSC when compared to the general population. Patients with HIV have a nearly 3-fold increased risk of developing primary NMSC as compared to the general population and a 44% increased risk of subsequent NMSC [49,50,51,52]. Additionally, the estimated standardized incidence rate for invasive NMSC is up to 82 times greater in patients who have received a kidney transplant when compared to non-transplanted patients [53]. Furthermore, immunosuppressed patients have an increased risk for rare types of NMSC, such as KS and MCC [54,55].
Lastly, the causal link between some risk factors and NMSC has been debated. Some evidence suggests a possible increased risk of NMSC in patients that are chronically exposed to certain photoreactive medications such as tetracycline, although further studies are needed to investigate these relationships [56]. Additionally, there are reported cases of NMSC occurring in the tattooed areas of the skin, but direct association between tattoos and skin cancers has not been established. As such, the FDA does not consider NMSCs complications of tattoo application [57]. Nevertheless, some still suggest that skin cancers should be included in the list of potential complications of tattooing given the actual incidence may be underreported [58].

2.2. Staging

BCCs rarely require staging given their minimal potential for metastasis. However, cutaneous SCC has a 4% annual incidence of metastasis, so staging is vital to its management and treatment [59,60,61]. The staging system of SCC is continuously being updated to meet current data. The staging system published in 2010 by the American Joint Committee on Cancers (AJCC) and the International Union Against Cancer (UICC) had some limitations. For example, T3/T4 tumors were reserved for bony invasion, which was very rare at 0.3–3.0%, while the rest were T1/T2 [62]. Thus, 70–86% of poor outcomes occurred in patients with T1/T2 tumors resulting in heterogeneous outcomes [62,63,64]. Whereas, in Brigham and Women’s Hospital (BWH) tumor staging, 5% of tumors were high stage but they accounted for 60% of poor outcomes, indicating superior homogeneity [62]. In 2017, the AJCC published the 8th edition of the Cancer Staging Manual. The staging of cutaneous SCC was revised to reflect recent evidence concerning high-risk clinicopathologic features to improve the overall staging system, which can be seen in Table 1, Table 2, Table 3 and Table 4 [63,65]. It should be noted that because the majority of SCCs of skin occur in the head and neck area, both of the AJCC’s 7th and 8th guidelines apply to tumors from the head and neck [63,64,65,66].

3. Prevention of NMSC

The incidence of NMSC has been increasing worldwide [1,2,3,4,67]. The rising incidence has been associated with several factors, including raised awareness in the general population and among physicians, an increased number of patients undergoing surgical treatment with confirmed histopathology instead of cryotherapy or electrodessication, improved registration, an aging population, and increased exposure to ultraviolent (UV) radiation [13,14,15,68]. These have led to the development of various skin cancer prevention measures and programs worldwide, highlighting the importance of skin cancer prevention.
Prevention strategies are divided into primary and secondary strategies. Primary prevention measures target behavioral changes to reduce sun exposure, reinforce the use of adequate sun protection and discourage intensive tanning, while the secondary measures facilitate early detection [4,69]. Public education is one of the key aspects of primary prevention, which is aimed to increase group-oriented awareness. It is known that among teen girls, correlating sun damage with premature wrinkling is an effective way to raise awareness [10,70]. In teen boys, emphasizing the protection of painful burns rather than the long-term risks of NMSC is more efficient [10,70]. Introduction of sun protection policy guidelines for schools and implementation of mass media campaigns of public education may also have significant impact on behavioral modifications [71,72]. Increasing evidence suggests that some pharmacologic agents may also be effective in prevention of NMSC. There is now strong evidence that vitamin D plays an important role in photocarcinogenesis and progression of NMSC. Hence, pharmacologic modulation by vitamin D, 1,25(OH)2D3 and its analogs represents a promising new strategy for prevention of NMSC [73]. Nicotinamide (Vitamin B3) is another agent that has been proven to decrease the incidence of NMSC, most likely by its ability to enhance DNA repair, modulate the inflammation produced by UVR and reduce UV induced immunosuppression [74,75]. Preventing and treating actinic keratosis (AKs) with minimal side effects and a low cost may be a promising alternative for skin cancer prevention in immunosuppressed patients [76].
Secondary prevention strategies include screening of high-risk populations, skin self-examination and physician surveillance, which aim to reduce morbidity and mortality by detecting cancer in its early stages [77].

4. Diagnosis and Types of Biopsy Techniques

The gold standard for diagnosing NMSC includes a thorough physical examination followed by conventional biopsy of the lesion for histopathologic examination [78,79]. A thorough lymph node exam is essential for diagnosis of SCC due to the risk for metastasis. Patients at high-risk for developing NMSC require special attention, particularly for individuals who work outside and thus have chronic exposure to UV radiation, as well as immunocompromised individuals. Some features, such as vaguely margined, white indurated, scar-like plaques, or the recently described “candle wax” sign may indicate an aggressive form of BCC [80,81,82,83].
When a suspicious lesion is identified, the diagnosis needs be confirmed with histopathologic analysis, which requires invasive intervention to obtain a tissue sample. There are several non-invasive medical technologies available that may be used to decide what lesions need to be biopsied as to potentially avoid unnecessary invasive procedures. Such technology includes: dermoscopy, confocal microscopy, cross-polarized light and fluorescence photography, as well as optical coherence tomography with high-frequency ultrasound [78]. There are also newer options, such as multiphoton microscopy and Raman spectroscopy that are emerging as state-of-the-art tools [78]. These adjunct screening tools may be used to assess skin lesions to find characteristic features of NMSC before proceeding with formal biopsy. The use of dermoscopy in the US is increasing with a recent survey showing up to 81% of US dermatologists using dermoscopy [84]. While it was mainly used for differentiation of melanocytic lesions, it is now widely utilized to screen superficial BCC and SCCs as well [85,86,87]. Most of these diagnostic modalities require additional training, which is one of the limiting factors for their use in clinic. Digitally stained multimodal confocal mosaic imaging was recently compared with standard frozen histopathologic specimens, thus showing 90% sensitivity and 79% specificity before training and 99% sensitivity and 93% specificity after training [88]. The longer processing time and the lack of reimbursement by the insurance companies are other limiting factors [89]. However, with the conduction of more research to justify their use, assignment of reimbursement values, development of more training courses for dermatologists, incorporating them into residency programs and continued improvement in speed and accuracy of these techniques will allow physicians to overcome some of the barriers and potentially increase the role of these technologies in the management of NMSC [89].

4.1. Biopsy Techniques

Despite tremendous advances in medical care and the shift to evidence-based medicine over the past several decades, there are no specific diagnostic guidelines for biopsy techniques that are specific to the lesion type or characteristics for one of the most common cancers worldwide. There are several types of biopsy techniques that are commonly used in the diagnosis of NMSC, yet no technique has been deemed the gold standard. These techniques include: excisional, incisional, shave, and punch biopsy. Although punch and shave biopsies are the most commonly performed techniques for initial sampling of NMSC [90], no well-powered studies in current literature or guidelines exist to help the physician decide which biopsy technique should be employed. Most studies are retrospective reviews and are not able to provide evidence-based proof that one technique is superior over the others, but rather demonstrate that each technique has advantages and disadvantages. It has been shown that although the intent of a shave biopsy is to obtain a diagnostic sample, 15–40% of patients who underwent secondary excisions for BCC had no residual cancer [91,92,93,94]. While some argue that those 15–40% of patients may have undergone unnecessary secondary procedures, final histopathologic examinations may be inaccurate. Kimyai-Asadi and Goldberg showed that bread-loafing at 4 mm intervals of elliptical excision specimens from facial basal cell carcinomas excised with 2 mm surgical margins is only 44% sensitive in detecting residual tumor at the surgical margins [95]. Another study demonstrated that 33% of cases with the initial biopsy results of actinic keratosis had additional findings on deep sections, including 13% SCC, 4% BCC, and 3% invasive SCC [96]. In the same context, a recent study brings up another critical point while questioning the value of reporting margin status in biopsies of NMSC. Based on their findings, 23% of cases with negative margins on initial biopsy demonstrated positive margins upon deeper-level examination [97] to further highlight the significance of proper interpretation of biopsy results when formulating a treatment plan.
With technology emerging in all aspects of the healthcare field, many look for new solutions to NMSC diagnosis. A recent study suggests that the reflectance confocal microscopy (RCM) may have a comparable accuracy to punch biopsy with regards to diagnosing and subtyping BCC [96,98,99]. It may also have a role in monitoring nonsurgical skin cancer therapies or assessment of tumor margins prior to surgery or intraoperatively [100,101,102,103].
In summary, there are currently no guidelines available to direct the biopsy techniques for the diagnosis of NMSCs. As a result, the clinician may decide what technique is the most appropriate in each scenario by considering not only the characteristics of the lesions, but also the intent of the biopsy. We are however hopeful that this issue will be addressed soon as the American Academy of Dermatology plans on releasing such guidelines in 2017 [104,105].

4.2. Low-Risk versus High-Risk Lesions

The current treatment of NMSC is driven by a risk assessment of the lesions. These assessments are based on the recurrence rates for BCC and recurrence and metastasis for SCC. According to NCCN 2017 guidelines, NMSC are classified as low- and high-risk lesions [106]. The lesions that are located on the trunk and extremities, excluding pretibia, hands, feet, nails, ankles, and are less than 20 mm in size are considered low-risk, whereas greater than 20 mm lesions are considered high-risk lesions. If the lesions are located on the cheeks, forehead, scalp, neck, and pretibia, the cutoff for size is 10 mm between the high and low-risk lesions. Other factors that differentiate the low and high-risk lesions include: well defined versus poorly defined borders, primary versus recurrent lesions, immunocompetent versus immunosuppressed, no history or history of RT at lesion site and no perineural involvement. Histologic subtypes also affect the recurrence and/or metastatic risk of the lesions such as nodular superficial BCCs and well or moderately differentiated SCCs are considered low-risk, whereas aggressive growth patterns for BCCs and poor differentiation for SCCs on histologic examination are indicative for high-risk lesions; see Table 5 for details [106].
RT-radiation therapy, L-trunk and extremities (excluding pretibia, hands, feet, nail units, and ankles), M-cheeks, forehead, scalp, neck, pretibia, H-central face, eyelids, eyebrows, periorbital, nose, lips, chin, mandible, preauricular and post auricular skin/sulci, temple, ear, genitalia, hands, feet.

4.3. The Role of Imaging

The majority of NMSCs can be successfully managed without imaging. Nevertheless, some SCCs may be locally invasive and have the potential for distant metastasis, while large or aggressive BCCs can invade surrounding anatomic structures. Therefore, high-risk tumors may require radiologic imaging for optimal management (Table 6). When there is concern for bony invasion, computed tomography (CT) should be employed [107,108]. Likewise, if there is concern for soft tissue or perineural involvement, magnetic resonance imaging (MRI) should be performed [107,108].

5. Treatment of NMSC: Overview of Surgical and Non-Surgical Treatment Modalities; NCCN Guidelines and Surgical Margins

NMSCs are considered curable diseases and the treatment mostly comprises of lesion removal. In addition, systemic and topical pharmacotherapy, cryotherapy (CT), photodynamic therapy (PDT), laser, and radiotherapy (RT) are also used [109,110,111,112,113]. A variety of techniques may be employed when removing NMSC. Common techniques include tangential shave removal, curettage, electrodessication, Mohs micrographic surgery (MMS), and standard surgical excision [114,115]. Many factors come into play when deciding treatment strategies including the patients’ overall condition and the goals of care. The current literature suggests that the treating physician’s specialty could also affect the technique utilized. For example, while some dermatology articles suggest that tangential shave removal of certain lesions provides an acceptable cure rate, Stewart et al. found an unacceptably high rate of residual BCC and SCC after shave biopsies [114,116]. This reflects the controversy surrounding type of biopsy employed previously in this article.
Concerns surrounding therapeutic non-surgical NMSC removal is detailed by Garcia-Cazana et al.; one of the main concerns of non-surgical treatments has been the development of resistance through a variety of mechanisms since these modalities usually require multiple administrations [117,118]. Despite the wide range of non-surgical treatment modalities that currently exist, surgery remains the mainstay of treatment for the majority of patients with NMSC and offers the highest cure rate [119,120].

5.1. Surgical Approach: Mohs Micrographic Surgery vs. Surgical Excision

It is generally agreed upon that surgical removal of NMSC offers the best overall outcome. A recent meta-analysis comparing the different treatment modalities for NMSC demonstrated that surgical excision had superior outcomes when compared to cryotherapy, photodynamic therapy, radiotherapy, 5-flurouracil (5-FU), and imiquimod in terms of complete lesion response, clearance of NMSC, and cumulative recurrence probabilities [109]. Additionally, Drew et al. compared MMS with non-surgical therapies (imiquimod, FU, ingenol mebutate, cryotherapy) for superficial BCC and SCCis, which demonstrated that both MMS and non-surgical therapies had high 5-year recurrence free survivals with MMS having a small advantage. Moreover, more patients would prefer to undergo MMS again due to the prolonged treatment course and pain associated with non-MMS treatments [11].
In a systemic review by Bath-Hextall et al., surgical excision was compared to MMS and demonstrated no difference in 30-month recurrence rates in the treatment of BCC [121]. Similarly, no difference was found in the recurrence risk following treatment of recurrent BCC removed by surgical excision versus MMS [122]. A recent meta-analysis by Lansbury et al. indicated similar results for cutaneous SCC, signifying no difference between recurrence risk of surgical excision and MMS [123]. Ghareeb et al. recently published an article comparing the outcome of MMS versus surgical excision for less common skin cancers in 15,121 patients identified through the National Cancer Data Base [124]. They showed only 8% were treated with MMS, however, the odds of achieving negative margins was 3.15 times higher in patients undergoing MMS when compared to all of the other surgical procedures combined when controlled for tumor size, location and histology. When MMS was compared with four different surgical techniques individually, it was still superior to primary excision and biopsy followed by narrow excision, however, no statistically significant difference was identified when comparing MMS with wide excision and re-excision with >1 cm margins [124].
The anatomic location of the tumor may pose unique challenges in its surgical excision. It should be noted that head and neck skin cancers are particularly challenging to treat given the difficulty in determining the amount of tissue that should be excised to provide adequate cancer-free margins with the best functional and cosmetic outcome. Moncrief et al. showed that the common practice of intraoperative frozen section analysis (IFSA) of margins had unacceptably high false negatives for head and neck BCC (28.7%) and SCC (27.5%). This resulted in the abandonment of IFSA in their practice and support for intraoperative MMS, even in advanced cases [125]. In light of this, Walker et al. investigated the utility of quenched activity-based probe imaging to discriminate cancerous versus normal skin tissue for NMSC [126]. They validated the activation of the probe with hematoxylin-eosin-confirmed cancerous tissue with 0.989 and 0.894 sensitivity and specificity, respectively. However, further studies are needed to confirm if this new, rapid, and easy to interpret technology would provide actual cost-effective increased cure rates by reducing re-excision and recurrence rates [126].
Although surgical excision remains the most commonly performed procedure in the treatment of NMSC, adequate surgical margins have not yet been defined. In an attempt to address this issue, Shel et al. suggested surgical margins for low and high-risk BCC and SCC based on a retrospective review of margins on 495 lesions as defined by MMS [127]. They concluded that lesions located on the face are best treated with MMS but in situations when standard surgical excision is the only option, a minimum 5 mm margin for low-risk lesions and about a 1 cm margin for high-risk lesions are required to completely excise the lesions in 95% of cases [127].
NCCN currently recommends standard excision as a primary treatment choice for low-risk BCCs that can be excised with 4 mm clinical margins as well as low-risk SCCs that can be excised with 4–6 mm clinical margins [106]. In case of positive margins, MMS or resection with complete margin assessment or standard re-excision in the trunk and extremity area are recommended [106]. In high-risk BCCs and local high-risk SCCs, MMS or resection with a complete margin assessment is recommended. Standard excision with wider surgical margins with linear or delayed repair may also be performed for high-risk lesions; however, there are no specific recommendations for margin size, and if margins are positive, then MMS or resection with complete margin assessment can be performed [106].

5.2. Curettage and Electrodessication (C&E), Cryosurgery

Curettage and electrodessication (C&E) is a commonly performed procedure to remove various skin lesions by scraping off the lesion with a curate followed by cauterization. It can be used for low-risk BCC and SCC, but it is not recommended for high-risk lesions due to an unacceptably high recurrence rate of up to 27% [128,129,130,131,132,133]. The overall efficacy of the C&E has been shown to be dependent on physicians’ experience as well. Silverman et al. showed decreased average recurrence rate of BCCs from 17.0% to 7.3%, when comparing the rates following procedures performed in the years of 1955–1963 and 1973–1982 by the same group, which was the attributed to physicians’ experience, specifically increased thoroughness in the technique [134]. Curettage alone may be used for selected low-risk lesions and can have similar cure rates (96% 5-year cure rate) but better healing compared to C&E [130]. Overall, C&E is considered a reasonable treatment choice for small low-risk NMSCs. It is also the least expensive and fastest method among all treatment methods of NMSCs [130,131].
Cryotherapy may also be considered for small, low-risk BCCs and SCCs, and can be combined with curettage, but it is not recommended for high-risk lesions [132,133]. It is associated with low cost, yet given the high overall recurrence rate of 7.5% following the treatment of primary lesions and 13% for recurrent lesions, it is rarely utilized [135,136].
According to current NCCN guidelines, C&E is recommended as a primary treatment choice for low-risk BCCs and local low-risk SCCs excluding terminal hair-bearing areas, such as the scalp, pubic and axillary region, beard area in men, and if adipose tissue is reached [106].

5.3. Photodynamic Therapy (PDT)

Photodynamic therapy (PDT) utilizes light, oxygen, and photosensitizers. 5-Aminolevulinic acid (5-ALA) and methyl 5-aminolevulinate (MAL) are commonly used pro-drugs for PDT, which are administered locally or systemically and convert to protoporphyrin IX (PPIX), which is then further converted to heme inside the cells. However, the PPIX conversion to heme is usually a slower process which results in its accumulation in cancer cells. Upon light absorption, the PPIX then undergoes excitation, resulting in production of oxygen free radicals, ultimately leading to cancer cell death. [137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160]. PDT has shown to be effective in treating superficial BCC, SCCis, and actinic keratosis (AK) with excellent cosmesis, though, it is currently not recommended for invasive SCC and aggressive BCC subtypes such as basosquamous or morphoeic infiltrating types [110,111]. A recent retrospective review demonstrated excellent response of superficial BCC to MAL-PDT and thick BCC to meta-tetrahydroxyphenylchlorin-PDT (mTHPC-PDT), with a 95% complete response rate after one round of PDT, which was maintained at 3-year follow-up and slightly decreased to 92% at 5-year follow-up [161]. PDT has also proven effective in reducing the incidence of AKs and SCCis in patients with organ transplant on long-term immunosuppressive therapy [110,111]. A randomized, multi-center study of 81 organ transplant recipients with 881 NMSCs (mainly AKs) as compared MAL-PDT to standard treatment (curettage, cryotherapy, surgery, or laser) and demonstrated significantly fewer new AKs in the MAL-PDT treated area at 15 months post-treatment [143]. These results indicate that pre-treatment of “field cancerization” in immunosuppressed patients with PDT is a promising alternative in terms of skin cancer prevention in immunosuppressed patients.
Significant research is being conducted to enhance the efficacy of PDT to treat more advanced stages of skin cancers. A recently developed microneedle-assisted PDT is aimed to deliver targeted therapy, which would allow treatment of deeper lesions [112,144]. In a small case series, the combination of a 630 nm laser with intralesional 5-aminolevulinic acid allowed treatment of deeper lesions [145]. A recent clinical trial studying the administration of 5-FU as a neoadjuvant for 5-aminolevulinic acid-PDT (ALA-PDT) has shown increased tumor selective protoporphyrin IX levels and enhanced cell death in a mouse mode [146]. Another clinical trial has compared the efficacy of ALA-PDT with surgery versus surgery alone for SCC of all stages, demonstrating a lower recurrence risk in the experimental group (16.6% versus 30.0%, p < 0.005) [147]. New agents are also developed with higher photosensitizing efficacy and better pharmacokinetics to provide higher tumor uptake and potentially long-term tumor cure. Patel et al. developed a near infrared bacteriochlorin 3 and the corresponding stereoisomers to be used in PDT with higher photosensitizing efficacy and limited skin phototoxicity when compared to porphyrin-based PDT [148].
PDT has also been studied as a potential modality for the prevention of NMSC. Studies demonstrated an equal preventative effect of daylight-PDT (dl-PDT) and conventional-PDT (c-PDT) in high-risk patients; however, all of the patients reported a burning sensation and pain during c-PDT while dl-PDT was reported to be almost pain free, which is why dl-PDT is preferred [149,150,151]. The authors postulate that PDT can induce premature senescence and kill senescent cells induced by itself [151]. A large retrospective review in a high volume dermatologic clinic investigated the development of SCC at one year following the PDT treatment of 1404 patients with AKs and 45 with NMSCs [152]. They found that 11% of patients developed SCC, while the rest remained SCC free at 1-year follow-up. Factors associated with developing SCC were older age, SCC history, Fitzpatrick skin-type 1, and sixty-minute or less incubation time, which is the time period between the administration of photosensitizing agent and exposure of the skin area to the light source. Therefore, it was concluded that PDT may be more effective in younger patients and with greater than 60 min of incubation time [152]. In addition, PDT has a tolerable side effect profile and low cost, which makes it more appealing in the treatment and prevention of skin cancers [153]. The main side effect is pain, which increases in intensity with the number of sessions and if the lesions are located in the head and neck area [153].

5.4. Laser Therapy

Treatment of NMSC with lasers causes light absorption by blood vessels of the targeted area, resulting in thermal distraction that then leads to tumor regression [154,155]. This targeted vascular photothermal destruction preserves the normal surrounding area and with treatment of dermatologic conditions, can lead to excellent cosmesis [155,156].
There are four major laser types used in the treatment of skin cancers: solid-state, diode, dye, and gas lasers [157]. A recent review article by Soleymani et al. discusses the different types of laser treatments in detail [158]. This study demonstrated that vascular targeting pulse dye lasers (PDL) have shown promising results in different studies with a complete clinical response in up to 95% of patients; nevertheless, poor results were also seen. Most incomplete responders were large tumors. Lower energy and small spot size also contributed to a poor cure rate. The 595 nm wavelength lasers were superior to the 585 nm wavelength lasers in terms of treating BCC. They concluded that the 595 nm wavelength vascular-targeted lasers had promising clinical efficacy in the treatment of BCC; however, data for consistent long-term outcome is not yet available. CO2 lasers allow very thin tissue ablation in the range of 20 µm with each pass [159,160,161,162,163]. Several studies have shown a different efficacy of CO2 lasers for treatment of BCC with 85–100% cure rates, excellent cosmetic outcomes, and minimal complications [158]. Lasers have been less commonly used and studied for the treatment of SCC, with only a handful of studies demonstrating the efficacy of CO2 lasers in treatment of SCCis. In the largest study of CO2 laser therapy, 44 of the 48 patients with SCCis were treated with CO2 lasers and had a total clearance rate of 97.7% and a recurrence rate of 6.8% at a mean 18-month follow-up [164].
Laser therapies have also been used in combination with PDT and the results have been mixed. One study investigated the efficacy of MAL-PDT with and without ablative (CO2) laser for the treatment of microinvasive SCC [165]. They found a greater efficacy rate when treated with the PDT-laser combination when compared to PDT alone at three months (84.2% versus 52.4%, p-0.03), and the differences in efficacy remained significant at 24 months. They also found a significantly lower recurrence rate in the PDT-laser group compared to the PDT alone group (12.5% versus 63.6%, p-0.006) [165]. In a case series, all patients with solitary superficial BCC treated with a combination of fractional CO2 laser and PDT achieved pathologically confirmed resolution of BCC with no serious adverse effects [152]. PDT combined with 2940 nm ER:YAG (solid-state) laser treatment has also shown improved relative efficacy in a few studies, demonstrating a lower recurrence risk of BCC and Bowen disease as compared to laser alone therapy, however, the esthetic outcomes were superior in laser alone treatment patients and the patients seem to prefer it due to its simplicity [157,166,167,168,169] Other studies dispute the benefits of PDT-laser combined therapy. One study compared the efficacy of PDT combined with pulse dye laser (PDL) with PDT alone for the treatment of 62 BCCs in 15 patients [170] and found no difference in complete tumor regression between the two groups. Additionally, this study found that the recurrence rate was higher in the combined treatment group [170].
Diode lasers, also called near-infrared lasers, function through temperature increases of target tissue and kill tumor cells directly. They have shown some efficacy in the treatment of small superficial SCC [157].
In summary, laser therapy has demonstrated promising results in the treatment of superficial NMSCs; however, the overall efficacy remains inferior compared to more traditional treatment modalities. It appears that the most beneficial aspects of laser treatment are the minimal side effects and excellent cosmetic outcome. While its role may be expanded in the future for the treatment of NMSCs, its use is currently limited. More long-term follow-up studies are needed to validate their efficacy and determine possible indications of laser therapy in the management of NMSCs.

5.5. Radiation Therapy

Although radiation therapy has shown control rates of 75–100% in early stage BCC and SCC, the role of radiotherapy has significantly decreased in the treatment of NMSCs with the emergence of MMS [113,171,172,173,174,175]. Now, it is mainly recommended as a primary treatment method if surgery is contraindicated, if lesions are located in cosmetically sensitive areas, or as an adjunct to surgery in patients with advanced disease [113].
There are several techniques of radiotherapy, each having advantages and disadvantages. A recent survey study completed by 16 members of American Brachytherapy Society indicated that the majority of them prefer brachytherapy over external beam radiation therapy given the shorter treatment course, conformity of treatment for irregular or curved targets and shallow dose deposition [176]. Many of them routinely use ultrasound to estimate the depth and lateral extension of the lesion before initiating treatment, which has shown to be helpful for more accurate treatment planning [176,177]. The advantage of superficial radiotherapy (SRT) is that the maximum dose is at skin’s surface, and there is less penetration through eye shields, thus making it easier to use around the eye [178]. It has shown to provide 93–100% tumor control depending on the lesion size, whereas electron beam radiation provided 72–88% tumor control in the same study [179]. Another study demonstrated a recurrence risk of 5% at five years in patients with low-risk NMSCs that are treated with SRT [180]. Electron beam radiotherapy (EBRT) has the advantage of treating superficial lesions without causing significant damage to deeper structures and may be useful when treating skin cancers over bony prominences or cartilaginous structures [181]. Interstitial brachytherapy is another subtype that involves the insertion of a radioactive catheter within the tumor bed and is used to treat skin cancers in difficult areas such as eyelids [182].
Some of the common complications of radiation therapy include alopecia, pigmentary changes, telangiectasias, fibrosis, atrophy, buccal mucositis, gingivitis, tooth loss, and a loss of salivary gland function. In addition, more severe complications such as soft tissue or bone necrosis, cataracts, conjunctival scarring, or eyelid deformity may occur [113,183].
NCCN currently recommends radiotherapy as a primary treatment modality for non-surgical candidates in the treatment of low- and high-risk BCCs and local low- and high-risk SCCs (in high-risk SCCs, RT may be supplemented with chemotherapy) [106]. However, it is often reserved for patients over 60 years of age due to concerns for long-term sequelae. Conversely, it is contraindicated in patients with genetic conditions predisposing to skin cancer and connective tissue diseases [106]. RT is also recommended in high-risk lesions in cases of positive margins after surgical excision and/or if a negative margin is unachievable with MMS. It may also be considered if there is extensive perineural or large nerve involvement [106]. NCCN recommends RT with or without chemotherapy in patients with SCCs with positive lymph nodes on FNA or core biopsy who are not surgical candidates or possible adjuvant therapy following surgical resection of lesions [107].

5.6. Chemotherapy and Immunotherapy

Systemic chemotherapy has an important role in the management of advanced NMSC. The term “advanced NMSC” usually refers to metastatic disease and/or inoperable lesions due to size and location that would otherwise cause unacceptable functional impairment if treated with excision [21,113]. Platinum based preparations such as cisplatin, are the most commonly used chemotherapeutic agents for NMSC; however, other agents, including cyclophosphamide, bleomycin, doxorubicin, methotrexate, and 5-FU, may also be used [23,24,25,26,27,28,29,30,31,121,122,184,185,186,187,188,189]. A recent publication claims that monotherapy of patients with stage I and II lip SCC via superficial temporal artery administration of bleomycin derivative peplomycin is a highly effective treatment method for achieving a cure in 70.8% of patients [113]. It was reported though, that 12.5% of patients developed interstitial pneumonia, 12.5% had occurrence of metastasis, and 16.7% had local recurrence [113].
Certain immune modulators and monoclonal antibodies have demonstrated promising results. In this concept, epidermal growth factor receptor (EGFR) appears to be of particular interest. EGFR is involved in the pathogenesis of SCC [190], hence its inhibitors, such as cetuximab and panitumumab, may be used in the treatment of SCC [23]. In one study, cetuximab has shown 69% efficacy in disease control rate at 6 weeks when administered as a single agent in patients with advanced SCC [191]. In another study, 95% of patients became operable following administration of cetuximab combined with a chemotherapeutic agent [192]. Cetuximab’s reasonable tolerability makes it a potential agent for neo-adjuvant therapy [23]. In contrast, panitumumab did not show a superior efficacy in combination with chemotherapy over chemotherapy alone in CONSERT-1 phase II trial or in combination with radiotherapy over chemotherapy alone in CONSERT-2 phase II trial [193,194]. In both studies, grade 3, grade 4, and serious adverse effects were more common in the group receiving panitumumab, which makes the future of this agent questionable in the treatment of NMSCs.
NCCN currently recommends chemotherapy as a possible supplement to radiation therapy in local, high-risk SCCs for patients who are not surgical candidates [106]. It is also recommended as a primary treatment concurrent with RT in patients with FNA/core biopsy positive lymph nodes who have inoperable diseases [106].

5.7. The Hedgehog Pathway Inhibitors

The ability of some antifungal agents to inhibit the Hedgehog (HH) pathway has also been explored in the treatment of NMSC. While itraconazole has shown some efficacy in exploratory phase II trial, the toxicity remains a major limiting factor in terms of its use in clinical practice. Posaconazole is another antifungal with better safety profile, though further validation is still needed [23,195].
Vismodegib is a newer agent that acts as an inhibitor of the sonic hedgehog (SHH) pathway and was approved for adult patients with advanced BCC in 2012 [21,196]. Despite some controversial reports of Vismodegib being associated with development of new SCC, it still appears to have great interest among physicians; however, its significant adverse effects pose a challenge [21,197,198,199]. Sonidegib is another SHH inhibitor recently approved for treatment of advanced BCC in the United States of America (USA) with slightly better efficacy than Vismodegib [200]. They are both currently recommended by NCCN to be considered in high-risk BCCs if positive margins are present following surgical excision and/or negative margins are not achievable with MMS [106].

5.8. Topical Agents

Topical agents may be used to treat NMSC in instances when patients are not candidates for standard approach. Factors that may favor the use of topical pharmacotherapy include extensive, multifocal or multiple tumors, indistinct lesion boundaries, localization in cosmetically sensitive or difficult to treat areas, and a history of hypertrophic scars and keloid [201]. There are several topical agents that are currently used in the treatment of NMSC, some of which are approved by FDA. Topical 5-FU has been approved for treatment of AK and superficial BCC for many years, although its use for treatment of SCCis is off-label [202,203,204]. Some studies suggest a 90–96% cure rate of AK and superficial BCC with topical 5-FU [205,206]. Imiquimod is another agent that is approved by FDA for treatment of AK and superficial BCC, while diclofenac and ingenol mebutate are FDA approved for AK only [202]. Currently, there are many other topical agents still in the investigational stage, including: resiquimod, piroxicam, calcium dobesilate, and potassium dobesilate containing formulations, betulinin acid and topical retinoids that may have some efficacy for the treatment of skin cancers [202].

6. Utility of Sentinel Lymph Node Biopsy (SLNB) and Regional Lymphadenectomy

Sentinel lymph node biopsy (SLNB) is usually not considered for BCC given the very rare chance of metastasis. In contrast, SLNB may play a significant role in the management of SCC. An association of positive SNL and poor prognosis of cutaneous SCC has been previously demonstrated in several studies, showing a 96% 5-year survival with no regional lymph node involvement [206,207,208,209,210,211,212,213,214,215]. The 5-year survival decreases to 72% with adequate treatment, and 25–35% with no treatment [207,208,209,210,211,212,213,214,215,216]. A recent study showed that patients with no micrometastasis to SNL developed no local and distant disease at an average follow-up period of 27.5 months [217]. Another recently published report indicated that positive results were seen in 13.4% of 364 SNLBs in patients with cutaneous SCC, which is similar to that of melanoma [218]. They also found that positive SNL was associated with poor prognosis; hence their findings further support the utility of SLNB in the management of SCC. However, while it appears that consensus exists in terms of positive rate of SNL and poor prognosis associated with it [219,220], many still question the utility of SNLB due to inadequate evidence that supports it actually improves the prognosis the disease. Maryuma et al. showed that despite an 18.4% positive rate of micrometastasis to SNL in patients who underwent SNLB, there was no difference in terms of metastasis-free and disease-specific survival rates between the groups who did and did not undergo SNLB, regardless of T-staging [221]. Silberstein et al. investigated the rate of metastasis to lymph nodes in 572 patients with 725 head and neck cutaneous T1 and T2 SCC with 1.09% and 5.46% positive results, respectively, and recommended no SNLB for clinically N0 patients based on these findings [222]. While there is no indication for SLNB in the current NCCN guidelines, some authors still recommend it given that SNLB may potentially improve patients’ prognosis if enough data is available [106,217].
When palpable regional lymph nodes are present or abnormal lymph nodes are identified on imaging studies, FNA or core biopsy is warranted [106]. In the case of positive results, excision of primary tumor and proper nodal dissection followed by chemoradiation therapy is a standard of care; however, prophylactic regional nodal dissection in clinically N0 disease still remains controversial [106,222].

7. Conclusions

Despite the emergence of novel non-surgical treatment modalities, surgical resection remains the most common treatment method for NMSCs with 4 mm clinical margins in low-risk BCCs and 4–6 mm in local low-risk SCCs. However, NCCN offers no clear guidelines in terms of margins in high-risk lesions. In high-risk BCCs and local high-risk SCCs, MMS, or surgical resection with complete margin assessment is recommended. RT is usually utilized for local lesions if the patient is not a surgical candidate or in metastatic disease with or without chemotherapy. Several topical agents, such as 5-FU and imiquimod, are FDA approved for superficial BCCs and AKs, while diclofenac and ingenol mebutate are FDA approved for AKs only. However, the off-label use of topical agents for treatment of SCC is also reported. PDT and laser therapy are relatively newer treatment modalities for NMSCs, with a reasonable success rate in low-risk lesions, though, more research is required to study their long-term outcomes, which may allow for the expansion of their utility in the management of skin cancers. Finally, the sonic hedgehog pathway inhibition is another emerging area of the treatment NMSCs with vismodegib and sonidegib already being approved for high-risk BCCs.
There appears to be no consensus in terms of recommended skin biopsy techniques for definitive diagnosis, as well as the role of sentinel lymph node biopsy. Though sentinel lymph node biopsy is warranted in cases of palpable lymphadenopathy, current data on regional lymph node dissection are sparse and more research is needed for any recommendation.

Author Contributions

Artur Fahradyan, Anna Howell, Erik Wolfswinkel, Michaela Tsuha and Parthiv Sheth made substantial contributions and design, and/or acquisition of data, and/or analysis and interpretation of data. Artur Fahradyan, Anna Howell, Erik Wolfswinkel and Alex K. Wong participated in drafting the article or revising it critically for important intellectual content. Alex K. Wong gave final approval of the version to be submitted and any revised version.

Conflicts of Interest

The authors declare no conflict of interest.

Financial Disclosure Statement

The authors have no financial interest to declare in relation to the content of this manuscript.

References

  1. Lomas, A.; Leonardi-Bee, J.; Bath-Hextall, F. A systematic review of worldwide incidence of nonmelanoma skin cancer. Br. J. Dermatol. 2012, 166, 1069–1080. [Google Scholar] [CrossRef] [PubMed]
  2. Rogers, H.W.; Weinstock, M.A.; Harris, A.R.; Hinckley, M.R.; Feldman, S.R.; Fleischer, A.B.; Coldiron, B.M. Incidence estimate of nonmelanoma skin cancer in the United States, 2006. Arch. Dermatol. 2010, 146, 283–287. [Google Scholar] [CrossRef] [PubMed]
  3. Leiter, U.; Eigentler, T.; Garbe, C. Epidemiology of skin cancer. Adv. Exp. Med. Biol. 2014, 810, 120–140. [Google Scholar] [PubMed]
  4. Apalla, Z.; Lallas, A.; Sotiriou, E.; Lazaridou, E.; Ioannides, D. Epidemiological trends in skin cancer. Dermatol. Pract. Concept. 2017, 7, 1–6. [Google Scholar] [CrossRef] [PubMed]
  5. Katalinic, A.; Kunze, U.; Schafer, T. Epidemiology of cutaneous melanoma and non-melanoma skin cancer in Schleswig-Holstein, Germany: Incidence, clinical subtypes, tumour stages and localization (epidemiology of skin cancer). Br. J. Dermatol. 2003, 149, 1200–1206. [Google Scholar] [CrossRef] [PubMed]
  6. Losquadro, W. Anatomy of the Skin and the Pathogenesis of Nonmelanoma Skin Cancer. Facial Plast. Surg. Clin. N. Am. 2017, 25, 283–289. [Google Scholar] [CrossRef] [PubMed]
  7. Eide, M.J.; Weinstock, M.A. Epidemiology of skin cancer. In Cancer of the Skin, 2nd ed.; Rigel, D.S., Robinson, J.K., Ross, M., Friedman, R.J., Cockerell, C.J., Lim, H.W., Stockfleth, E., Kirkwood, J.M., Eds.; Elsevier: Amsterdam, The Netherlands, 2011; pp. 44–55. [Google Scholar]
  8. Wollina, U. Carcinosarcoma of skin (sarcomatoid carcinoma)—A rare non-melanoma skin cancer (Case Review). Georgian Med. News 2017, 263, 7. [Google Scholar]
  9. Barton, V.; Armeson, K.; Hampras, S.; Ferris, L.K.; Visvanathan, K.; Rollison, D.; Alberg, A.J. Nonmelanoma skin cancer and risk of all-cause and cancer-related mortality: A systematic review. Arch. Dermatol. Res. 2017, 309, 243–251. [Google Scholar] [CrossRef] [PubMed]
  10. Lewis, K.; Weinstock, M. Nonmelanoma Skin Cancer Mortality (1988–2000). Arch. Dermatol. 2004, 140, 837–842. [Google Scholar] [CrossRef] [PubMed]
  11. Guy, G.; Machlin, S.; Ekwueme, D.; Yabroff, K. Prevalence and Costs of Skin Cancer Treatment in the U.S., 2002−2006 and 2007−2011. Am. J. Prev. Med. 2015, 48, 183–187. [Google Scholar] [CrossRef] [PubMed]
  12. Urbach, F.; Forbes, P.D.; Davies, R.E.; Berger, D. Cutaneous photobiology: Past, present and future. J. Investig. Dermatol. 1976, 67, 209–224. [Google Scholar] [CrossRef] [PubMed]
  13. Fears, T.; Scotto, J.; Schneiderman, M. Mathematical models of age and ultraviolet effects on the incidence of skin cancer among whites in the United States. Am. J. Epidemiol. 1977, 105, 420–427. [Google Scholar] [CrossRef] [PubMed]
  14. Fears, T.; Scotto, J.; Schneiderman, M. The Authors Reply. Am. J. Epidemiol. 1978, 107, 260–262. [Google Scholar] [CrossRef]
  15. Armstrong, B.; Cust, A. Sun exposure and skin cancer, and the puzzle of cutaneous melanoma. Cancer Epidemiol. 2017, 48, 147–156. [Google Scholar] [CrossRef] [PubMed]
  16. Hampras, S.; Locke, F.; Chavez, J.; Patel, P.S.; Giuliano, A.R.; Miller, K.; Gheit, T.; Tommasino, M.; Rollison, D.E. Prevalence of cutaneous viral infections in incident cutaneous squamous cell carcinoma detected among chronic lymphocytic leukemia and hematopoietic stem cell transplant patients. Leuk. Lymphoma 2017, 1–7. [Google Scholar] [CrossRef] [PubMed]
  17. Hiesse, C.; Rieu, P.; Kriaa, F.; Larue, J.R.; Goupy, C.; Neyrat, N.; Charpentier, B. Malignancy after renal transplantation: Analysis of incidence and risk factors in 1700 patients followed during a 25-year period. Transplant. Proc. 1997, 29, 831–833. [Google Scholar] [CrossRef]
  18. Greenberg, J.N.; Zwald, F.O. Management of Skin Cancer in Solid-organ Transplant Recipients: A Multidisciplinary Approach. Dermatol. Clin. 2011, 29, 231–241. [Google Scholar] [CrossRef] [PubMed]
  19. Penn, I. Post-transplant malignancy: The role of immunosuppression. Drug Saf. 2000, 23, 101–113. [Google Scholar] [CrossRef] [PubMed]
  20. Pollard, J.D.; Hanasono, M.M.; Mikulec, A.A.; Le, Q.T.; Terris, D.J. Head and neck cancer in cardiothoracic transplant recipients. Laryngoscope 2000, 110, 1257–1261. [Google Scholar] [CrossRef] [PubMed]
  21. Marzuka, A.G.; Book, S.E. Basal cell carcinoma: Pathogenesis, epidemiology, clinical features, diagnosis, histopathology, and management. Yale J. Biol. Med. 2015, 88, 167–179. [Google Scholar] [PubMed]
  22. Fogel, A.; Sarin, K.; Teng, J. Genetic diseases associated with an increased risk of skin cancer development in childhood. Curr. Opin. Pediatr. 2017, 29, 426–433. [Google Scholar] [CrossRef] [PubMed]
  23. Amaral, T.; Garbe, C. Non-melanoma skin cancer: New and future synthetic drug treatments. Exp. Opin. Pharmacother. 2017, 18, 689–699. [Google Scholar] [CrossRef] [PubMed]
  24. Perkins, J.L.; Liu, Y.; Mitby, P.A.; Neglia, J.P.; Hammond, S.; Stovall, M.; Meadows, A.T.; Hutchinson, R.; Dreyer, Z.E.; Robinson, L.L.; Mertens, A.C. Nonmelanoma skin cancer in survivors of childhood and adolescent cancer: A report from the childhood cancer survivor study. J. Clin. Oncol. 2005, 23, 3733–3741. [Google Scholar] [CrossRef] [PubMed]
  25. Karagas, M.R.; Nelson, H.H.; Zens, M.S.; Linet, M.; Stukel, T.A.; Spencer, S.; Applebaum, K.M.; Mott, L.; Mabuchi, K. Squamous cell and basal cell carcinoma of the skin in relation to radiation therapy and potential modification of risk by sun exposure. Epidemiol. Camb. Mass 2007, 18, 776–784. [Google Scholar] [CrossRef] [PubMed]
  26. Watt, T.C.; Inskip, P.D.; Stratton, K.; Smith, S.A.; Kry, S.F.; Sigurdson, A.L.; Stovall, M.; Leisenning, W.; Robison, L.L.; Mertens, A.C. Radiation-related risk of basal cell carcinoma: A report from the Childhood Cancer Survivor Study. J. Natl. Cancer Inst. 2012, 104, 1240–1250. [Google Scholar] [CrossRef] [PubMed]
  27. Schwartz, J.L.; Kopecky, K.J.; Mathes, R.W.; Leisenring, W.M.; Friedman, D.L.; Deeg, H.J. Basal cell skin cancer after total-body irradiation and hematopoietic cell transplantation. Radiat. Res. 2009, 171, 155–163. [Google Scholar] [CrossRef] [PubMed]
  28. Khalesi, M.; Whiteman, D.C.; Tran, B.; Kimlin, M.G.; Olsen, C.M.; Neale, R.E. A meta-analysis of pigmentary characteristics, sun sensitivity, freckling and melanocytic nevi and risk of basal cell carcinoma of the skin. Cancer Epidemiol. 2013, 37, 534–543. [Google Scholar] [CrossRef] [PubMed]
  29. Bernat Garcia, J.; Morales Suarez-Varela, M.; Vilata, J.J.; Marguina, A.; Pallardo, L.; Crespo, J. Risk factors for non-melanoma skin cancer in kidney transplant patients in a Spanish population in the Mediterranean region. Acta Derm. Venereol. 2013, 93, 422–427. [Google Scholar] [CrossRef] [PubMed]
  30. Zilinska, Z.; Sersenova, M.; Chrastina, M.; Breza, J., Sr.; Bena, L.; Baltesova, T.; Jurcina, A.; Roland, R.; Lackova, E.; Cellar, M.; Laca, L.; Dedinska, I. Occurrence of malignancies after kidney transplantation in adults: Slovak multicenter experience. Neoplasma 2017, 64, 311–317. [Google Scholar] [CrossRef] [PubMed]
  31. Parren, L.J.; Frank, J. Hereditary tumour syndromes featuring basal cell carcinomas. Br. J. Dermatol. 2011, 165, 30–34. [Google Scholar] [CrossRef] [PubMed]
  32. Karagas, M.R.; Nelson, H.H.; Sehr, P.; Waterboer, T.; Stukel, T.A.; Andrew, A.; Green, A.C.; Bavinck, J.N.; Perry, A.; Spencer, S.; et al. Human papillomavirus infection and incidence of squamous cell and basal cell carcinomas of the skin. J. Natl. Cancer Inst. 2006, 98, 389–395. [Google Scholar] [CrossRef] [PubMed]
  33. Patel, A.S.; Karagas, M.R.; Perry, A.E.; Nelson, H.H. Exposure profiles and human papillomavirus infection in skin cancer: An analysis of 25 genus beta-types in a population-based study. J. Investig. Dermatol. 2008, 128, 2888–2893. [Google Scholar] [CrossRef] [PubMed]
  34. Nikolaou, V.; Stratigos, A.J.; Tsao, H. Hereditary nonmelanoma skin cancer. Semin. Cutan. Med. Surg. 2012, 31, 204–210. [Google Scholar] [CrossRef] [PubMed]
  35. Cerdan Santacruz, C.; Diaz del Arco, C.; Rubio Herrera, M.; Sanchez-Pernaute, A.; Torres Garcia, A. Squamous Cell Carcinoma of the Peristomal Skin of a Gastrostomy. J. Wound Ostomy Cont. Nurs. 2017, 44, 384–386. [Google Scholar] [CrossRef] [PubMed]
  36. Oh, P.; Gill, K.; Lynch, L.; Cowles, R. Primary squamous cell carcinoma arising at a gastrostomy tube site. J. Pediatr. Surg. 2011, 46, 756–758. [Google Scholar] [CrossRef] [PubMed]
  37. Arons, M.S.; Lynch, J.R.; Lewis, S.R.; Blocker, T.G., Jr. Scar tissue carcinoma, part I: A clinical study with special reference to burn scar carcinoma. Ann. Surg. 1965, 161, 170–188. [Google Scholar] [CrossRef] [PubMed]
  38. Ramanujam, P.; Venkatesh, K.S. An unusual case of squamous cell carcinoma arising at the stomal site: Case report and review of the literature. J. Gastrointest. Surg. 2002, 6, 630–631. [Google Scholar] [CrossRef]
  39. Losanoff, J.E.; Sochaki, P.; Khoury, N.; Levi, E.; Salwen, W.A.; Basson, M.D. Squamous cell carcinoma complicating chronic suppurative hydradenitis. Am. Surg. 2011, 77, 1449–1453. [Google Scholar] [PubMed]
  40. Lavogiez, C.; Delaporte, E.; Darras-Vercambre, S.; Martin De Lassalle, E.; Castillo, C.; Mirabel, X.; Laurent, F.; Patenotre, P.; Gheit, T.; Talmant, J.C.; et al. Clinicopathological study of 13 cases of squamous cell carcinoma complicating hidradenitis suppurativa. Dermatology 2010, 220, 147–153. [Google Scholar] [CrossRef] [PubMed]
  41. Cruickshank, A.H.; Gaskell, E. Jean-Nicolas Marjolin: Destined to be forgotten? Med. Hist. 1963, 7, 383–384. [Google Scholar] [CrossRef] [PubMed]
  42. Metwally, I.; Roshdy, A.; Saleh, S.; Ezzat, M. Epidemiology and predictors of recurrence of Marjolin’s ulcer: Experience from Mansoura Universityxs. Ann. R. Coll. Surg. Engl. 2017, 99, 245–249. [Google Scholar] [CrossRef] [PubMed]
  43. Bartos, V.; Kullova, M. Basal Cell Carcinoma Multiplicity–A Retrospective Analysis of 899 Biopsy-proven Patients from a Single Institute. Klinicka Onkol. 2017, 30, 197–201. [Google Scholar] [CrossRef] [PubMed]
  44. Ramachandran, S.; Fryer, A.A.; Smith, A.; Lear, J.; Bowers, B.; Jones, P.W.; Strange, R.C. Cutaneous basal cell carcinomas: Distinct host factors are associated with the development of tumors on the trunk and on the head and neck. Cancer 2001, 92, 354–358. [Google Scholar] [CrossRef]
  45. Karagas, M.R.; Greenberg, E.R. Unresolved issues in the epidemiology of basal cell and squamous cell skin cancer. In Skin Cancer: Mechanisms and Human Relevance; Mukhtar, H., Ed.; CRC Press: Boca Raton, FL, USA, 1995; pp. 79–86. [Google Scholar]
  46. Kiiski, V.; de Vries, E.; Flohil, S.C.; Bill, M.J.; Hofman, A.; Stricker, B.H.; Nijsten, T. Risk factors for single and multiple basal cell carcinomas. Arch. Dermatol. 2010, 146, 848–855. [Google Scholar] [CrossRef] [PubMed]
  47. Flohil, S.C.; Koljenovic, S.; de Haas, E.R.; Overbeek, L.I.; de Vries, E.; Nijsten, T. Cumulative risks and rates of subsequent basal cell carcinomas in Netherlands. Br. J. Dermatol. 2011, 165, 874–881. [Google Scholar] [CrossRef] [PubMed]
  48. Lovatt, T.J.; Lear, J.T.; Bastrilles, J.; Wong, C.; Griffiths, C.E.; Samarasinghe, V.; Roebuck, J.; Ramachandran, S.; Smith, A.G.; Jones, P.W.; et al. Associations between ultraviolet radiation, basal cell carcinoma site and histology, host characteristics, and rate of development of further tumors. J. Am. Acad. Dermatol. 2005, 52, 468–473. [Google Scholar] [CrossRef] [PubMed]
  49. Weistenhofer, W.; Hiller, J.; Drexler, H.; Kiesel, J. Retrospective evaluation of exposure to natural UV radiation: Experiences with the online UV history tool in a field study. JDDG J. Dtsch. Dermatol. Ges. 2017, 15, 610–619. [Google Scholar] [CrossRef] [PubMed]
  50. Schaart, F.; Garbe, C.; Orfanos, C. Disappearance of the ozone layer and skin cancer: Attempt at risk assessment. Hautarzt 1993, 44, 63–68. [Google Scholar] [PubMed]
  51. Silverberg, M.J.; Leyden, W.; Warton, E.M.; Quesenberry, C.P., Jr.; Engels, E.A.; Asgari, M.M. HIV infection status, immunodeficiency, and the incidence of non-melanoma skin cancer. J. Natl. Cancer Inst. 2013, 105, 350–360. [Google Scholar] [CrossRef] [PubMed]
  52. Asgari, M.; Ray, G.; Quesenberry, C.; Katz, K.; Silverberg, M. Association of Multiple Primary Skin Cancers with Human Immunodeficiency Virus Infection, CD4 Count, and Viral Load. JAMA Dermatol. 2017, 153, 892–896. [Google Scholar] [CrossRef] [PubMed]
  53. Moloney, F.; Comber, H.; O’Lorcain, P.; O’Kelly, P.; Conlon, P.; Murphy, G. A population-based study of skin cancer incidence and prevalence in renal transplant recipients. Br. J. Dermatol. 2006, 154, 498–504. [Google Scholar] [CrossRef] [PubMed]
  54. Lindelof, B.; Sigurgeirsson, B.; Gabel, H.; Stern, R. Incidence of skin cancer in 5356 patients following organ transplantation. Br. J. Dermatol. 2000, 143, 513–519. [Google Scholar] [PubMed]
  55. Buell, J.; Trofe, J.; Hanaway, M.; Beebe, T.M.; Gross, T.G.; Alloway, R.R.; First, M.R.; Woodle, E.S. Immunosuppression and merkel cell cancer. Transpl. Proc. 2002, 34, 1780–1781. [Google Scholar] [CrossRef]
  56. Vakharia, P.; Nardone, B.; Schlosser, B.; Lee, D.; Serrano, L.; West, D. Chronic exposure to tetracyclines and subsequent diagnosis for non-melanoma skin cancer in a large Midwestern U.S. patient population. J. Eur. Acad. Dermatol. Venereol. 2017. [Google Scholar] [CrossRef] [PubMed]
  57. FDA. Tattoos & Permanent Makeup: Fact Sheet. Available online: https://www.fda.gov/cosmetics/productsingredients/products/ucm108530.htm (accessed on 24 July 2017).
  58. Junqueira, A.; Wanat, K.; Farah, R. Squamous neoplasms arising within tattoos: Clinical presentation, histopathology and management. Clin. Exp. Dermatol. 2017, 42, 601–606. [Google Scholar] [CrossRef] [PubMed]
  59. Burton, K.; Ashack, K.; 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] [PubMed]
  60. Ting, P.T.; Kasper, R.; Arlette, J.P. Metastatic basal cell carcinoma: Report of two cases and literature review. J. Cutan. Med. Surg. 2005, 9, 10–15. [Google Scholar] [CrossRef] [PubMed]
  61. Toll, A.; Margalef, P.; Masferrer, E.; Ferrandiz-Pulido, C.; Gimeno, J.; Pujol, R.M.; Bigas, A.; Espinosa, L. Active nuclear IKK correlates with metastatic risk in cutaneous squamous cell carcinoma. Arch. Dermatol. Res. 2015, 307, 721–729. [Google Scholar] [CrossRef] [PubMed]
  62. Karia, P.; Jambusaria-Pahlajani, A.; Harrington, D.; Murphy, G.; Qureshi, A.; Schmults, C. Evaluation of American Joint Committee on Cancer, International Union Against Cancer, and Brigham and Women’s Hospital Tumor Staging for Cutaneous Squamous Cell Carcinoma. J. Clin. Oncol. 2014, 32, 327–334. [Google Scholar] [CrossRef] [PubMed]
  63. Motaparthi, K.; Kapil, J.; Velazquez, E. Cutaneous Squamous Cell Carcinoma. Adv. Anatomic Pathol. 2017, 24, 171–194. [Google Scholar] [CrossRef] [PubMed]
  64. Edge, S.B.; Byrd, D.R.; Compton, C.C.; Fritz, A.G.; Greene, F.L.; Trotti, A. Cutaneous Squamous Cell Carcinoma and Other Cutaneous Carcinomas American Joint Committee on Cancer. In AJCC Cancer Staging Handbook, 7th ed.; Springer: New York, NY, USA, 2010; Chapter 29. [Google Scholar]
  65. Califano, J.A.; Lydiatt, W.M.; Nehal, K.S.; O’Sullivan, B.; Schmults, C.; Seethala, R.R.; Weber, R.S.; Shah, J.P. Chapter 15: Cutaneous Squamous Cell Carcinoma of the Head and Neck AJCC Cancer Staging Manual, 8th ed.; Springer: New York, NY, USA, 2017; pp. 171–181. [Google Scholar]
  66. McCormack, C. Differences in age and body site distribution of the histological subtypes of basal cell carcinoma. A possible indicator of differing causes. Arch. Dermatol. 1997, 133, 593–596. [Google Scholar] [CrossRef] [PubMed]
  67. Muzic, J.; Schmitt, A.; Wright, A.; Alniemi, D.T.; Zubair, A.S.; Olazagasti Lourido, J.M.; Sosa Seda, I.M.; Weaver, A.L.; Baum, C.L. Incidence and Trends of Basal Cell Carcinoma and Cutaneous Squamous Cell Carcinoma. Mayo Clin. Proc. 2017, 92, 890–898. [Google Scholar] [CrossRef] [PubMed]
  68. Verkouteren, J.; Ramdas, K.; Wakkee, M.; Nijsten, T. Epidemiology of basal cell carcinoma: Scholarly review. Br. J. Dermatol. 2017, 177, 359–372. [Google Scholar] [CrossRef] [PubMed]
  69. Olson, A.L.; Gaffney, C.A.; Starr, P.; Dietrich, A.J. The impact of an appearance-based educational intervention on adolescent intention to use sunscreen. Health Educ. Res. 2008, 23, 763–769. [Google Scholar] [CrossRef] [PubMed]
  70. Reeder, A.I.; Jopson, J.A.; Gray, A. Sun protection policies and practices in New Zealand primary schools. N. Z. Med. J. 2012, 125, 70–82. [Google Scholar] [PubMed]
  71. Doran, C.M.; Ling, R.; Byrnes, J.; Crane, M.; Shakeshaft, A.P.; Searles, A.; Perez, D. Benefit Cost Analysis of Three Skin Cancer Public Education Mass-Media Campaigns Implemented in New South Wales, Australia. PLoS ONE 2016, 11, e0147665. [Google Scholar] [CrossRef] [PubMed]
  72. Robinson, J.K.; Mallett, K.A. The duty to inspect the skin and counsel those at risk to develop melanoma. JAMA 2009, 301, 1702–1704. [Google Scholar] [CrossRef] [PubMed]
  73. Reichrath, J.; Saternus, R.; Vogt, T. Endocrine actions of vitamin D in skin: Relevance for photocarcinogenesis of non-melanoma skin cancer, and beyond. Mol. Cell. Endocrinol. 2017, 453, 96–102. [Google Scholar] [CrossRef] [PubMed]
  74. Chen, A.; Martin, A.; Choy, B.; Fernandez-Penas, P.; Dalziell, R.A.; McKenzie, C.A.; Scolyer, R.A.; Dhillon, H.M.; Vardy, J.L.; Kricker, A.; et al. A Phase 3 Randomized Trial of Nicotinamide for Skin-Cancer Chemoprevention. N. Engl. J. Med. 2015, 373, 1618–1626. [Google Scholar] [CrossRef] [PubMed]
  75. Minocha, R.; Damian, D.; Halliday, G. Melanoma and non-melanoma skin cancer chemoprevention: A role for nicotinamide? Photodermatol. Photoimmunol. Photomed. 2017. [Google Scholar] [CrossRef] [PubMed]
  76. Drago, F.; Ciccarese, G.; Cogorno, L.; Calvi, C.; Marsano, L.A.; Parodi, A. Prevention of non-melanoma skin cancers with nicotinamide in transplant recipients: A case-control study. Eur. J. Dermatol. 2017, 27, 382–385. [Google Scholar] [PubMed]
  77. Cummings, S.R.; Trip, M.K.; Herrmann, N.B. Approaches to the prevention and control of skin cancer. Cancer Metastasis Rev. 1997, 16, 309–327. [Google Scholar] [CrossRef]
  78. Malvehy, J.; Pellacani, G. Dermoscopy, Confocal Microscopy and other Non-invasive Tools for the Diagnosis of Non-Melanoma Skin Cancers and Other Skin Conditions. Acta Derm. Venereol. 2017. [Google Scholar] [CrossRef] [PubMed]
  79. Werner, R.N.; Stockfleth, E.; Connolly, S.M.; Correia, O.; Erdmann, R.; Foley, P.; Gupta, A.K.; Jacobs, A.; Kerl, H.; Lim, H.W.; et al. Evidence- and consensus-based (S3) Guidelines for the treatment of actinic keratosis–International League of Dermatological Societies in cooperation with the European Dermatology Forum–short version. J. Eur. Acad. Dermatol. Venereol. 2015, 29, 2069–2079. [Google Scholar] [CrossRef] [PubMed]
  80. Dourmishev, L.; Rusinova, D.; Botev, I. Clinical variants, stages, and management of basal cell carcinoma. Indian Dermatol. Online J. 2013, 4, 12. [Google Scholar] [CrossRef] [PubMed]
  81. Walling, H.; Fosko, S.; Geraminejad, P.; Whitaker, D.; Arpey, C. Aggressive basal cell carcinoma: Presentation, pathogenesis, and management. Cancer Metastasis Rev. 2004, 23, 389–402. [Google Scholar] [CrossRef] [PubMed]
  82. Rubin, A.I.; Chen, E.H.; Ratner, D. Basal-cell carcinoma. N. Engl. J. Med. 2005, 353, 2262–2269. [Google Scholar] [CrossRef] [PubMed]
  83. O’Donnell, B.; Duarte, C. A prospective evaluation of the candle wax sign: A visual clue to diagnose aggressive basal cell carcinoma. J. Am. Acad. Dermatol. 2017, 77, 163–164. [Google Scholar] [CrossRef] [PubMed]
  84. Murzaku, E.; Hayan, S.; Rao, B. Methods and rates of dermoscopy usage: A cross-sectional survey of US dermatologists stratified by years in practice. J. Am. Acad. Dermatol. 2014, 71, 393–395. [Google Scholar] [CrossRef] [PubMed]
  85. Katz, B.J.; Oliviero, M.; Rabinovitz, H. Dermoscopy and its impact on skin cancer diagnostics. J. Drugs Dermatol. 2010, 9, 129–130. [Google Scholar] [PubMed]
  86. Felder, S.; Rabinovitz, H.; Oliviero, M.; Kopf, A. Dermoscopic differentiation of a superficial basal cell carcinoma and squamous cell carcinoma in situ. Dermatol. Surg. 2006, 32, 423–425. [Google Scholar]
  87. Amjadi, M.; Coventry, B.; Greenwood, A.M.J. Non-Invasive Tools for Improving Diagnosis of Non-Melanoma Skin Cancer: A Review. Int. J. Plast. Surg. 2010, 7, 1–5. [Google Scholar]
  88. Mu, E.; Lewin, J.; Stevenson, M.; Meehan, S.; Carucci, J.; Gareau, D. Use of Digitally Stained Multimodal Confocal Mosaic Images to Screen for Nonmelanoma Skin Cancer. JAMA Dermatol. 2016, 152, 1335. [Google Scholar] [CrossRef] [PubMed]
  89. Que, S.; Grant-Kels, J.; Longo, C.; Pellacani, G. Basics of Confocal Microscopy and the Complexity of Diagnosing Skin Tumors. Dermatol. Clin. 2016, 34, 367–375. [Google Scholar] [CrossRef] [PubMed]
  90. Russell, E.; Carrington, P.; Smoller, B. Basal cell carcinoma: A comparison of shave biopsy versus punch biopsy techniques in subtype diagnosis. J. Am. Acad. Dermatol. 1999, 41, 69–71. [Google Scholar] [CrossRef]
  91. Gurunluoglu, R.; Kubek, E.; Arton, J.; Olsen, A.; Bronsert, M. Nonpersistence of Basal Cell Carcinoma after Diagnostic Shave Biopsy. Ann. Plast. Surg. 2015, 74, 695–698. [Google Scholar] [CrossRef] [PubMed]
  92. Alcalay, J.; Alkalay, R.; Hazaz, B. Residual skin cancer after preoperative biopsy: Evaluation by Mohs micrographic surgery. Int. J. Dermatol. 2004, 43, 456–458. [Google Scholar] [CrossRef] [PubMed]
  93. Alcalay, J.; Alkalay, R. Histological evaluation of residual basal cell carcinoma after shave biopsy prior to Mohs micrographic surgery. J. Eur. Acad. Dermatol. Venereol. 2010, 25, 839–841. [Google Scholar] [CrossRef] [PubMed]
  94. Grelck, K.; Sukal, S.; Rosen, L.; Suciu, G. Incidence of Residual Nonmelanoma Skin Cancer in Excisions after Shave Biopsy. Dermatol. Surg. 2013, 39, 374–380. [Google Scholar] [CrossRef] [PubMed]
  95. Kimyai-Asadi, A.; Goldberg, L.; Jih, M. Accuracy of serial transverse cross-sections in detecting residual basal cell carcinoma at the surgical margins of an elliptical excision specimen. J. Am. Acad. Dermatol. 2005, 53, 468–473. [Google Scholar] [CrossRef] [PubMed]
  96. Roozeboom, M.; Mosterd, K.; Winnepenninckx, V.; Nelemans, P.; Kelleners-Smeets, N. Agreement between histological subtype on punch biopsy and surgical excision in primary basal cell carcinoma. J. Eur. Acad. Dermatol. Venereol. 2012, 27, 894–898. [Google Scholar] [CrossRef] [PubMed]
  97. Schnebelen, A.; Gardner, J.; Shalin, S. Margin Status in Shave Biopsies of Nonmelanoma Skin Cancers: Is It Worth Reporting? Arch. Pathol. Lab. Med. 2016, 140, 678–681. [Google Scholar] [CrossRef] [PubMed]
  98. Sun, M.; Wu, A.; Huilgol, S.; Selva, D. Accuracy of Biopsy in Subtyping Periocular Basal Cell Carcinoma. Ophthalmic Plast. Reconstr. Surg. 2015, 31, 449–451. [Google Scholar] [CrossRef] [PubMed]
  99. Kadouch, D.; Leeflang, M.; Elshot, Y.; Longo, C.; Ulrich, M.; van der Wal, A.C.; Wolkerstorfer, A.; Bekkenk, M.W.; de Rie, M.A. Diagnostic accuracy of confocal microscopy imaging versus punch biopsy for diagnosing and subtyping basal cell carcinoma. J. Eur. Acad. Dermatol. Venereol. 2017, 31, 1641–1648. [Google Scholar] [CrossRef] [PubMed]
  100. Que, S.; Grant-Kels, J.; Rabinovitz, H.; Oliviero, M.; Scope, A. Application of Handheld Confocal Microscopy for Skin Cancer Diagnosis. Dermatol. Clin. 2016, 34, 469–475. [Google Scholar] [CrossRef] [PubMed]
  101. Cinotti, E.; Perrot, J.L.; Labeille, B.; Douchet, C.; Mottet, N.; Cambazard, F. Laser photodynamic treatment for in situ squamous cell carcinoma of the glans monitored by reflectance confocal microscopy. Australas. J. Dermatol. 2014, 55, 72–74. [Google Scholar] [CrossRef] [PubMed]
  102. Kai, A.C.; Richards, T.; Coleman, A.; Mallipeddi, R.; Barlow, R.; Craythorne, E.E. Five-year recurrence rate of lentigo maligna after treatment with imiquimod. Br. J. Dermatol. 2016, 174, 165–168. [Google Scholar] [CrossRef] [PubMed]
  103. Chen, C.S.; Sierra, H.; Cordova, M.; Rajadhyaksha, M. Confocal microscopy-guided laser ablation for superficial and early nodular basal cell carcinoma: A promising surgical alternative for superficial skin cancers. JAMA Dermatol. 2014, 150, 994–998. [Google Scholar] [CrossRef] [PubMed]
  104. Phelps, R.; Lebwohl, M. Biopsy Techniques. JAMA Dermatol. 2014, 150, 12. [Google Scholar] [CrossRef] [PubMed]
  105. American Academy of Dermatology|Association. Clinical Guidelines. Available online: https://www.aad.org/practicecenter/quality/clinical-guidelines#undefined (accessed on 20 July 2017).
  106. National Comprehensive Cancer Network. Available online: https://www.nccn.org/professionals/physician_gls/pdf/nmsc_blocks.pdf (accessed on 18 July 2017).
  107. Humphreys, T.; Shah, K.; Wysong, A.; Lexa, F.; MacFarlane, D. The role of imaging in the management of patients with nonmelanoma skin cancer: When is Imaging Necessary? J. Am. Acad. Dermatol. 2017, 76, 591–607. [Google Scholar] [CrossRef] [PubMed]
  108. MacFarlane, D.; Shah, K.; Wysong, A.; Wortsman, X.; Humphreys, T. The role of imaging in the management of patients with nonmelanoma skin cancer: Diagnostic Modalities and Applications. J. Am. Acad. Dermatol. 2017, 76, 579–588. [Google Scholar] [CrossRef] [PubMed]
  109. Lv, R.; Sun, Q. A Network Meta-Analysis of Non-Melanoma Skin Cancer (NMSC) Treatments: Efficacy and Safety Assessment. J. Cell. Biochem. 2017, 118, 3686–3695. [Google Scholar] [CrossRef] [PubMed]
  110. Griffin, L.; Lear, J. Photodynamic Therapy and Non-Melanoma Skin Cancer. Cancers 2016, 8, 98. [Google Scholar] [CrossRef] [PubMed]
  111. Cohen, D.; Lee, P. Photodynamic Therapy for Non-Melanoma Skin Cancers. Cancers 2016, 8, 90. [Google Scholar] [CrossRef] [PubMed]
  112. Hamdan, I.; Donnelly, R. Microneedle-assisted photodynamic therapy: Delivery of a NIR photosensitiser for the treatment of skin cancers. Photodiagnosis Photodyn. Ther. 2017, 17, A63. [Google Scholar] [CrossRef]
  113. Cheraghi, N.; Cognetta, A.; Goldberg, D. Radiation Therapy in Dermatology: Non-Melanoma Skin Cancer. J. Drugs Dermatol. 2017, 16, 464–469. [Google Scholar] [PubMed]
  114. Abramson, A.; Krasny, M.; Goldman, G. Tangential Shave Removal of Basal Cell Carcinoma. Dermatol. Surg. 2013, 39, 387–392. [Google Scholar] [CrossRef] [PubMed]
  115. Taheri, A.; Mansoori, P.; Laffer, M.; Feldman, S. Tangential Shave Removal of Basal Cell Carcinoma. Dermatol. Surg. 2013, 39, 1945–1946. [Google Scholar] [CrossRef] [PubMed]
  116. Stewart, C.; Garlick, J.; Mcmullin, J.; Siddigi, F.; Crombie, C.; Rockwell, W.B.; Gociman, B. Surgical Excision of Non–Melanoma Skin Cancer in an Elderly Veteran’s Affairs Population. Plast. Reconstr. Surg. Glob. Open 2014, 2, e277. [Google Scholar] [CrossRef] [PubMed]
  117. Gracia-Cazana, T.; Gonzalez, S.; Gilaberte, Y. Resistance of Nonmelanoma Skin Cancer to Nonsurgical Treatments. Part I: Topical Treatments. Actas Dermo-Sifiliogr. 2016, 107, 730–739. [Google Scholar] [CrossRef]
  118. Gracia-Cazana, T.; Salazar, N.; Zamarron, A.; Mascaraque, M.; Lucena, S.; Juarranz, Á. Resistance of Nonmelanoma Skin Cancer to Nonsurgical Treatments. Part II: Photodynamic Therapy, Vismodegib, Cetuximab, Intralesional Methotrexate, and Radiotherapy. Actas Dermo-Sifiliogr. 2016, 107, 740–750. [Google Scholar] [CrossRef] [PubMed]
  119. Telfer, N.R.; Colver, G.B.; Morton, C.A. Guidelines for the management of basal cell carcinoma. Br. J. Dermatol. 2008, 159, 35–48. [Google Scholar] [CrossRef] [PubMed]
  120. Motley, R.; Kersey, P.; Lawrence, C. Multiprofessional guidelines for the management of the patient with primary cutaneous squamous cell carcinoma. Br. J. Dermatol. 2002, 146, 18–25. [Google Scholar] [CrossRef] [PubMed]
  121. Bath-Hextall, F.J.; Perkins, W.; Bong, J.; Williams, H.C. Interventions for basal cell carcinoma of the skin. Cochrane Database Syst. Rev. 2007. [Google Scholar] [CrossRef]
  122. Smeets, N.W.; Krekels, G.A.; Ostertag, J.U.; Essers, B.A.; Dirksen, C.D.; Nieman, F.H.; Neuman, H.A. Surgical excision vs. Mohs’ micrographic surgery for basal-cell carcinoma of the face: Randomised controlled trial. Lancet 2004, 364, 1766–1772. [Google Scholar] [CrossRef]
  123. Lansbury, L.; Bath-Hextall, F.; Perkins, W.; Stanton, W.; Leonardi-Bee, J. Interventions for nonmetastatic squamous cell carcinoma of the skin: Systematic review and pooled analysis of observational studies. BMJ 2013, 347, f6153. [Google Scholar] [CrossRef] [PubMed]
  124. Ghareeb, E.; Dulmage, B.; Vargo, J.; Balasubramani, G.; Beriwal, S. Underutilization of Mohs Micrographic Surgery for Less Common Cutaneous Malignancies in the United States. Dermatol. Surg. 2016, 42, 653–662. [Google Scholar] [CrossRef] [PubMed]
  125. Moncrieff, M.; Shah, A.; Igali, L.; Garioch, J. False-negative rate of intraoperative frozen section margin analysis for complex head and neck nonmelanoma skin cancer excisions. Clin. Exp. Dermatol. 2015, 40, 834–838. [Google Scholar] [CrossRef] [PubMed]
  126. Walker, E.; Mann, M.; Honda, K.; Vidimos, A.; Schluchter, M.D.; Straight, B.; Bogyo, M.; Popkin, D.; Basilion, J.P. Rapid visualization of nonmelanoma skin cancer. J. Am. Acad. Dermatol. 2017, 76, 209–216. [Google Scholar] [CrossRef] [PubMed]
  127. Schell, A.; Russell, M.; Park, S. Suggested Excisional Margins for Cutaneous Malignant Lesions Based on Mohs Micrographic Surgery. JAMA Facial Plast. Surg. 2013, 15, 337. [Google Scholar] [CrossRef] [PubMed]
  128. McDaniel, W.E. Therapy for basal cell epitheliomas by curettage only: Further study. Arch. Dermatol. 1983, 119, 901–903. [Google Scholar] [CrossRef] [PubMed]
  129. Reymann, F. 15 years’ experience with treatment of basal cell carcinomas of the skin with curettage. Acta Derm. Venereol. Suppl. Stockh 1985, 120, 56–59. [Google Scholar] [PubMed]
  130. Barlow, J.O.; Zalla, M.J.; Kyle, A.; DiCaudo, D.J.; Lim, K.K.; Yiannias, J.A. Treatment of basal cell carcinoma with curettage alone. J. Am. Acad. Dermatol. 2006, 54, 1039–1045. [Google Scholar] [CrossRef] [PubMed]
  131. Kauvar, A.; Cronin, T., Jr.; Roenigk, R.; Hruza, G.; Bennett, R. Consensus for Nonmelanoma Skin Cancer Treatment; basal cell carcinoma, including a cost analysis of treatment methods. Dermatol. Surg. 2015, 41, 550–571. [Google Scholar] [CrossRef] [PubMed]
  132. Kauvar, A.; Arpey, C.; Hruza, G.; Olbricht, S.; Bennett, R. Consensus for Nonmelanoma Skin Cancer Treatment, Part II; Squamous Cell Carcinoma, Including a Cost Analysis of Treatment Methods. Dermatol. Surg. 2015, 41, 1214–1240. [Google Scholar] [CrossRef] [PubMed]
  133. Blixt, E.; Nelsen, D.; Stratman, E. Recurrence rates of aggressive histologic types of basal cell carcinoma after treatment with electrodesiccation and curettage alone. Dermatol. Surg. 2013, 39, 719–725. [Google Scholar] [CrossRef] [PubMed]
  134. Silverman, M.K.; Kopf, A.W.; Grin, C.M.; Bart, R.S.; Levenstein, M.J. Recurrence rates of treated basal cell carcinomas. Part 2: Curettage-electrodesiccation. J. Dermatol. Surg. Oncol. 1991, 17, 720–726. [Google Scholar] [CrossRef] [PubMed]
  135. Rowe, D.E.; Carroll, R.J.; Day, C.L., Jr. Mohs surgery is the treatment of choice for recurrent (previously treated) basal cell carcinoma. J. Dermatol. Surg. Oncol. 1989, 15, 424–431. [Google Scholar] [CrossRef] [PubMed]
  136. Rowe, D.E.; Carroll, R.J.; Day, C.L., Jr. Long-term recurrence rates in previously untreated (primary) basal cell carcinoma: Implications for patient follow-up. J. Dermatol. Surg. Oncol. 1989, 15, 315–328. [Google Scholar] [CrossRef] [PubMed]
  137. Kennedy, J.C.; Pottier, R.H.; Pross, D.C. Photodynamic therapy with endogenous protoporphyrin IX: Basic principles and present clinical experience. J. Photochem. Photobiol. B Biol. 1990, 6, 143–148. [Google Scholar] [CrossRef]
  138. Grant, W.E.; Hopper, C.; Speight, P.M.; Macrobert, A.J.; Brown, S.G. Photodynamic therapy of malignant and premalignant lesions in patients with ‘field cancerization’ of the oral cavity. J. Laryngol. Otol. 1993, 107, 1140–1145. [Google Scholar] [CrossRef] [PubMed]
  139. Grant, W.E.; Speight, P.M.; Hopper, C.; Brown, S.G. Photodynamic therapy: An effective, but non-selective treatment for superficial cancers of the oral cavity. Int. J. Cancer 1997, 71, 937–942. [Google Scholar] [CrossRef]
  140. Lou, P.J.; Jager, H.R.; Jones, L.; Theodossy, T.; Brown, S.G.; Hopper, C. Interstitial photodynamic therapy as salvage treatment for recurrent head and neck cancer. Br. J. Cancer 2004, 91, 441–446. [Google Scholar] [CrossRef] [PubMed]
  141. Jager, H.R.; Taylor, M.N.; Theodossy, T.; Hopper, C. MR imaging-guided interstitial photodynamic laser therapy for advanced head and neck tumors. AJNR Am. J. Neuroradiol. 2005, 26, 1193–1200. [Google Scholar] [PubMed]
  142. Jerjes, W.; Upile, T.; Betz, C.S.; el Maaytah, M.; Abbas, S.; Wright, A.; Hopper, C. The application of photodynamic therapy in the head and neck. Dent. Update Lond. 2007, 34, 478. [Google Scholar] [CrossRef]
  143. Wennberg, A.M.; Keohane, S.; Lear, J.T.; Jemec, G.; Mørk, C.; Christensen, E.; Kapp, A.; Sølvsten, H.; Talm, T.; Berne, B.; et al. Results from a 15-month update of a multicentre study of methyl aminolaevulinate photodynamic therapy in immunocompromised organ transplant recipients with nonmelanoma skin cancer. Br. J. Dermatol. 2006, 155, 57. [Google Scholar]
  144. Jain, A.; Lee, C.; Gill, H. 5-Aminolevulinic acid coated microneedles for photodynamic therapy of skin tumors. J. Control. Release 2016, 239, 72–81. [Google Scholar] [CrossRef] [PubMed]
  145. Suarez-Valladares, M.; Rodriguez-Prieto, M.; Serra-Llusa, R. Penetration of 630 nm laser and 5-aminolevulinic acid in tissue with intralesional photodynamic therapy. Photodiagnosis Photodyn. Ther. 2016, 16, 166–168. [Google Scholar] [CrossRef] [PubMed]
  146. Anand, S.; Rollakanti, K.; Brankov, N.; Brash, D.; Hasan, T.; Maytin, E. Fluorouracil Enhances Photodynamic Therapy of Squamous Cell Carcinoma via a p53-Independent Mechanism that Increases Protoporphyrin IX levels and Tumor Cell Death. Mol. Cancer Ther. 2017, 16, 1092–1101. [Google Scholar] [CrossRef] [PubMed]
  147. Peng, J.; Feng, W.; Luo, X.; Wang, T.; Xiang, W.; Dai, Y.; Zhu, J.; Zheng, J. A Clinical Trial Using Attrition Combined with 5-Aminolevulinic Acids Based Photodynamic Therapy in Treating Squamous Cell Carcinoma. Med. Sci. Monit. 2017, 23, 1347–1354. [Google Scholar] [CrossRef] [PubMed]
  148. Patel, N.; Pera, P.; Joshi, P.; Dukh, M.; Tabaczynski, W.A.; Siters, K.E.; Kryman, M.; Cheruku, R.R.; Durrani, F.; Missert, J.R.; et al. Highly Effective Dual-Function Near-Infrared (NIR) Photosensitizer for Fluorescence Imaging and Photodynamic Therapy (PDT) of Cancer. J. Med. Chem. 2016, 59, 9774–9787. [Google Scholar] [CrossRef] [PubMed]
  149. Sotiriou, E.; Apalla, Z.; Vrani, F.; Lazaridou, E.; Vakirlis, E.; Lallas, A.; Ionnides, D. Daylight photodynamic therapy vs. Conventional photodynamic therapy as skin cancer preventive treatment in patients with faces and scalp cancerization: An intra-individual comparison study. J. Eur. Acad. Dermatol. Venereol. 2017. [Google Scholar] [CrossRef] [PubMed]
  150. Apalla, Z.; Sotiriou, E.; Chovarda, E.; Lefaki, I.; Devliotou-Panagiotidou, D.; Ioannides, D. Skin cancer: Preventive photodynamic therapy in patients with face and scalp cancerization. A randomized placebo-controlled study. Br. J. Dermatol. 2010, 162, 171–175. [Google Scholar] [CrossRef] [PubMed]
  151. Grigalavicius, M.; Juraleviciute, M.; Kwitniewski, M.; Juzeniene, A. The influence of photodynamic therapy with 5-aminolevulinic acid on senescent skin cancer cells. Photodiagnosis Photodyn. Ther. 2017, 17, 29–34. [Google Scholar] [CrossRef] [PubMed]
  152. Orbuch, D.E.; Penn, L.; Bloom, B.S.; Brauer, J.A.; Shin, D.B.; Greenbaum, J.; Bernstein, L.J.; Weiss, E.T.; Anolik, R.T.; Geronemus, R.G. The Photodynamic Therapy Experience of a High Volume Laser and Dermatologic Surgery Center. J. Drugs Dermatol. 2016, 15, 1420–1426. [Google Scholar] [PubMed]
  153. Blanco, K.C.; Inada, N.M.; Silva, A.P.; Stringasci, M.D.; Buzza, H.H.; Ramirez, D.P.; Salvio, A.G.; Moriyama, L.T.; Kurachi, C.; Bagnato, V.S. A Multicenter Clinical Study of Expected and Unexpected Side Reactions during and after Skin Cancer Treatment by Photodynamic Therapy. Skinmed 2017, 15, 113–118. [Google Scholar] [PubMed]
  154. Anderson, R. Lasers for Dermatology and Skin Biology. J. Investig. Dermatol. 2013, 133, E21–E23. [Google Scholar] [CrossRef] [PubMed]
  155. Jalian, H.; Avram, M.; Stankiewicz, K.; Shofner, J.; Tannous, Z. Combined 585 nm pulsed-dye and 1064 nm Nd:YAG lasers for the treatment of basal cell carcinoma. Lasers Surg. Med. 2013, 46, 1–7. [Google Scholar] [CrossRef] [PubMed]
  156. Anderson, R.R.; Parrish, J.A. Selective photothermolysis: Precise microsurgery by selective absorption of pulsed radiation. Science 1983, 220, 524–527. [Google Scholar] [CrossRef] [PubMed]
  157. Mirza, F.; Khatri, K. The use of lasers in the treatment of skin cancer: A review. J. Cosmet. Laser Ther. 2017. [Google Scholar] [CrossRef] [PubMed]
  158. Soleymani, T.; Abrouk, M.; Kelly, K. An Analysis of Laser Therapy for the Treatment of Nonmelanoma Skin Cancer. Dermatol. Surg. 2017, 43, 615–624. [Google Scholar] [CrossRef] [PubMed]
  159. Zachary, C.B.; Rofagha, R. Laser therapy. In Dermatology, 3rd ed.; Bolognia, J.L., Jorizzo, J.L., Schaffer, J.V., Eds.; Mosby: London, UK, 2012. [Google Scholar]
  160. Omi, T.; Numano, K. The role of the CO2 laser and fractional CO2 laser in dermatology. Laser Ther. 2014, 23, 49–60. [Google Scholar] [CrossRef] [PubMed]
  161. Humphreys, T.R.; Malhotra, R.; Scharf, M.J.; Marcus, S.M.; Starkus, L.; Calegari, K. Treatment of superficial basal cell carcinoma and squamous cell carcinoma in situ with a high-energy pulsed carbon dioxide laser. Arch. Dermatol. 1998, 134, 1247–1252. [Google Scholar] [CrossRef] [PubMed]
  162. Trelles, M.; David, L.; Rigau, J. Penetration depth of ultrapulsed carbon dioxide laser in human skin. Dermatol. Surg. 1996, 22, 863–865. [Google Scholar] [CrossRef] [PubMed]
  163. Adams, E.L.; Price, N.M. Treatment of basal-cell carcinomas with a carbon-dioxide laser. J. Dermatol. Surg. Oncol. 1979, 5, 803–806. [Google Scholar] [CrossRef] [PubMed]
  164. Martinez-Gonzalez, M.C.; Pozo, J.D.; Paradela, S.; Fernández-Jorge, B.; Fernández-Torres, R.; Fonseca, E. Bowen’s disease treated by carbon dioxide laser. A series of 44 patients. J. Dermatol. Treat. 2008, 19, 293–299. [Google Scholar] [CrossRef] [PubMed]
  165. Choi, S.; Kim, K.; Song, K. Effect of Methyl Aminolevulinate Photodynamic Therapy With and Without Ablative Fractional Laser Treatment in Patients with Microinvasive Squamous Cell Carcinoma. JAMA Dermatol. 2017, 153, 289. [Google Scholar] [CrossRef] [PubMed]
  166. Smucler, R.; Vlk, M. Combination of Er:YAG laser and photodynamic therapy in the treatment of nodular basal cell carcinoma. Lasers Surg. Med. 2008, 40, 153–158. [Google Scholar] [CrossRef] [PubMed]
  167. Ko, D.Y.; Kim, K.H.; Song, K.H. A randomized trial comparing methyl aminolaevulinate photodynamic therapy with and without Er:YAG ablative fractional laser treatment in Asian patients with lower extremity Bowen disease: Results from a 12-month follow-up. Br. J. Dermatol. 2014, 170, 165–172. [Google Scholar] [CrossRef] [PubMed]
  168. Smucler, R.; Kriz, M.; Lippert, J.; Vlk, M. Ultrasound guided ablative-laser assisted photodynamic therapy of basal cell carcinoma (US-aL-PDT). Photomed. Laser Surg. 2012, 30, 200–205. [Google Scholar] [CrossRef] [PubMed]
  169. Choi, S.H.; Kim, K.H.; Song, K.H. Er:YAG ablative fractional laser-primed photodynamic therapy with methyl aminolevulinate as an alternative treatment option for patients with thin nodular basal cell carcinoma: 12-month follow-up results of a randomized, prospective, comparative trial. J. Eur. Acad. Dermatol. Venereol. 2016, 30, 783–788. [Google Scholar] [CrossRef] [PubMed]
  170. Carija, A.; Puizina-Ivic, N.; Vukovic, D.; Miric Kovacevic, L.; Capkun, V. Single treatment of low-risk basal cell carcinomas with pulsed dye laser-mediated photodynamic therapy (PDL-PDT) compared with photodynamic therapy (PDT): A controlled, investigator-blinded, intra-individual prospective study. Photodiagnosis Photodyn. Ther. 2016, 16, 60–65. [Google Scholar] [CrossRef] [PubMed]
  171. Kwan, W.; Wilson, D.; Moravan, V. Radiotherapy for locally advanced basal cell and squamous cell carcinomas of the skin. Int. J. Radiat. Oncol. Biol. Phys. 2004, 60, 406e411. [Google Scholar] [CrossRef] [PubMed]
  172. Locke, J.; Karimpour, S.; Young, G.; Lockett, M.A.; Perez, C.A. Radiotherapy for epithelial skin cancer. Int. J. Radiat. Oncol. Biol. Phys. 2001, 51, 748–755. [Google Scholar] [CrossRef]
  173. Schulte, K.W.; Lippold, A.; Auras, C.; Bramkamp, G.; Breitkopf, C.; Elsmann, H.J.; Habenicht, E.M.; Jasnoch, V.; Muller-Pannes, H.; Rupprecht, R.; et al. Soft X-ray therapy for cutaneous basal cell and squamous cell carcinomas. J. Am. Acad. Dermatol. 2005, 53, 993–1001. [Google Scholar] [CrossRef] [PubMed]
  174. Van Hezewijk, M.; Creutzberg, C.L.; Putter, H.; Chin, A.; Schneider, I.; Hoogeveen, M.; Willemze, R.; Marijnen, C.A. Efficacy of a hypofractionated schedule in electron beam radiotherapy for epithelial skin cancer: Analysis of 434 cases. Radiother. Oncol. 2010, 95, 245–249. [Google Scholar] [CrossRef] [PubMed]
  175. Guix, B.; Finestres, F.; Tello, J.; Palma, C.; Martinez, A.; Guix, J.; Guix, R. Treatment of skin carcinomas of the face by high-dose-rate brachytherapy and custom-made surface molds. Int. J. Radiat. Oncol. Biol. Phys. 2000, 47, 95–102. [Google Scholar] [CrossRef]
  176. Likhacheva, A.; Devlin, P.; Shirvani, S.; Barker, C.A.; Beron, P.; Bhatnagar, A.; Doggett, S.W.; Hochman, L.; Hsu, C.; Kasper, M.; et al. Skin surface brachytherapy: A survey of contemporary practice patterns. Brachytherapy 2017, 16, 223–229. [Google Scholar] [CrossRef] [PubMed]
  177. Goyal, U.; Kim, Y.; Tiwari, H.; Witte, R.; Stea, B. A pilot study of ultrasound-guided electronic brachytherapy for skin cancer. J. Contemp. Brachyther. 2015, 5, 374–380. [Google Scholar] [CrossRef] [PubMed]
  178. Mendenhall, W.M.; Amdur, R.J.; Hinerman, R.W.; Cognetta, A.B.; Mendenhall, N. Radiotherapy for cutaneous squamous and basal cell carcinomas of the head and neck. Laryngoscope 2009, 119, 1994–1999. [Google Scholar] [CrossRef] [PubMed]
  179. Lovett, R.D.; Perez, C.A.; Shapiro, S.J.; Garcia, D.M. External Irradiation of epithelial skin cancer. Int. J. Radiat. Oncol. Biol. Phys. 1990, 19, 235–242. [Google Scholar] [CrossRef]
  180. Cognetta, A.B.; Howard, B.M.; Heaton, H.P.; Stoddard, E.R.; Hong, H.G.; Green, H.W. Superficial X-ray in the treatment of basal and squamous cell carcinomas: A viable option in select patients. J. Am. Acad. Dermatol. 2012, 67, 1235–1241. [Google Scholar] [CrossRef] [PubMed]
  181. Miller, R.A.; Spittle, M.F. Electron beam therapy for difficult cutaneous basal and squamous cell carcinoma. Br. J. Dermatol. 1982, 106, 429–435. [Google Scholar] [CrossRef] [PubMed]
  182. Mareco, V.; Bujor, L.; Abrunhosa-Branquinho, A.N.; Ferreira, M.R.; Ribeiro, T.; Vasconcelos, A.L.; Ferreira, C.R.; Jorge, M. Interstitial high-dose-rate brachytherapy in eyelid cancer. Brachytherapy 2015, 14, 554–564. [Google Scholar] [CrossRef] [PubMed]
  183. Cho, M.; Gordon, L.; Remblelak, A.; Woo, T.C. Utility of radiotherapy for treatment of basal cell carcinoma: A review. Br. J. Dermatol. 2014, 171, 968–973. [Google Scholar] [CrossRef] [PubMed]
  184. Pfeiffer, P.; Hansen, O.; Rose, C. Systemic cytotoxic therapy of basal cell carcinoma. A review of the literature. Eur. J. Cancer 1990, 26, 73–77. [Google Scholar] [CrossRef]
  185. Carneiro, B.A.; Watkin, W.G.; Mehta, U.K.; Brockstein, B.E. Metastatic basal cell carcinoma: Complete response to chemotherapy and associated pure red cell aplasia. Cancer Investig. 2006, 24, 396–400. [Google Scholar] [CrossRef] [PubMed]
  186. Khandekar, J.D. Complete response of metastatic basal cell carcinoma to cisplatin chemotherapy: A report on two patients. Arch. Dermatol. 1990, 126, 1660. [Google Scholar] [CrossRef] [PubMed]
  187. Wysong, A.; Aasi, S.Z.; Tang, J.Y. Update on metastatic basal cell carcinoma: A summary of published cases from 1981 through 2011. JAMA Dermatol. 2013, 149, 615–616. [Google Scholar] [CrossRef] [PubMed]
  188. Espeli, V.; Ruegg, E.; Hottinger, A.F.; Modarressi, A.; Dietrich, P.Y. Weekly multi-agent chemotherapy (CMF-b) for advanced non-melanoma skin cancer. Anticancer Res. 2016, 36, 2359–2364. [Google Scholar] [PubMed]
  189. Sadek, H.; Azli, N.; Wendling, J.L.; Cvitkovic, E.; Rahal, M.; Mamelle, G.; Guillaume, J.C.; Armand, J.P.; Avril, M.F. Treatment of advanced squamous cell carcinoma of the skin with cisplatin, 5-fluorouracil, and bleomycin. Cancer 1990, 66, 1692–1696. [Google Scholar] [CrossRef]
  190. Krahn, G.; Leiter, U.; Kaskel, P.; Udart, M.; Utikal, J.; Bezold, G.; Peter, R.U. Coexpression patterns of EGFR, HER2, HER3 and HER4 in non-melanoma skin cancer. Eur. J. Cancer 2001, 37, 251–259. [Google Scholar] [CrossRef]
  191. 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] [PubMed]
  192. Reigneau, M.; Robert, C.; Routier, E.; Mamelle, G.; Moya-Plana, A.; Tomasic, G.; Mateus, C. Efficacy of neoadjuvant cetuximab alone or with platinum salt for the treatment of unresectable advanced nonmetastatic cutaneous squamous cell carcinomas. Br. J. Dermatol. 2015, 173, 527–534. [Google Scholar] [CrossRef] [PubMed]
  193. Mesia, R.; Henke, M.; Fortin, A.; Minn, H.; Yunes Ancona, A.C.; Cmelak, A.; Markowitz, A.B.; Hotte, S.J.; Singh, S.; Chan, A.T.; et al. Chemoradiotherapy with or without panitumumab in patients with unresected, locally advanced squamous-cell carcinoma of the head and neck (CONCERT-1): A randomised, controlled, open-label phase 2 trial. Lancet Oncol. 2015, 16, 208–220. [Google Scholar] [CrossRef]
  194. Giralt, J.; Trigo, J.; Nuyts, S.; Ozsahin, M.; Skladowski, K.; Hatoum, G.; Daisne, J.F.; Yunes Ancona, A.C.; Cmelak, A.; Mesia, R.; et al. Panitumumab plus radiotherapy versus chemoradiotherapy in patients with unresected, locally advanced squamous-cell carcinoma of the head and neck (CONCERT-2): A randomized, controlled, open-label phase 2 trial. Lancet Oncol. 2015, 16, 221–232. [Google Scholar] [CrossRef]
  195. Kim, D.J.; Kim, J.; Spaunhurst, K.; Montoya, J.; Khodosh, R.; Chandra, K.; Fu, T.; Gilliam, A.; Molgo, M.; Beachy, P.A.; et al. Open-label, exploratory Phase II trial of oral itraconazole for the treatment of basal cell carcinoma. J. Clin. Oncol. 2014, 32, 745–751. [Google Scholar] [CrossRef] [PubMed]
  196. Bakshi, A.; Chaudhary, S.; Rana, M.; Elmets, C.; Athar, M. Basal cell carcinoma pathogenesis and therapy involving hedgehog signaling and beyond. Mol. Carcinog. 2017. [Google Scholar] [CrossRef] [PubMed]
  197. Alarcon, I.; Pasquali, P.; Malvehy, J.; Puig, S. Tumor regrowth and development of keratinocytic neoplasms in patients under smoothened inhibition: In vivo assessment with reflectance confocal microscopy. Skin Res. Technol. 2016, 23, 283–288. [Google Scholar] [CrossRef] [PubMed]
  198. Gjersvik, P. Study on the risk of cutaneous squamous cell carcinoma after vismodegib therapy for Basal cell carcinoma. JAMA Dermatol. 2016, 152, 1172. [Google Scholar] [CrossRef] [PubMed]
  199. Zargari, O.; Azimi, S.; Geranmayeh, S. Inoperable infiltrative basal cell carcinoma successfully treated with vismodegib. Dermatol. Ther. 2017, 30, e12509. [Google Scholar] [CrossRef] [PubMed]
  200. Odom, D.; Mladsi, D.; Purser, M.; Kaye, J.A.; Palaka, E.; Charter, A.; Jensen, J.A.; Sellami, D. A Matching-Adjusted Indirect Comparison of Sonidegib and Vismodegib in Advanced Basal Cell Carcinoma. J. Skin Cancer 2017, 2017, 1–7. [Google Scholar] [CrossRef] [PubMed]
  201. Micali, G.; Lacarrubba, F.; Nasca, M.R.; Ferraro, S.; Schwartz, R.A. Topical pharmacotherapy for skin cancer: Part II. Clinical applications. J. Am. Acad. Dermatol. 2014, 70, 979.e1–979.e12. [Google Scholar] [CrossRef] [PubMed]
  202. Micali, G.; Lacarrubba, F.; Nasca, M.R.; Schwartz, R.A. Topical pharmacotherapy for skin cancer: Part I. Pharmacology. J. Am. Acad. Dermatol. 2014, 70, 965.e1–965.e12. [Google Scholar] [CrossRef] [PubMed]
  203. Bargman, H.; Hochman, J. Topical treatment of Bowen’s disease with 5-fluorouracil. J. Cutan. Med. Surg. 2003, 7, 101–105. [Google Scholar] [CrossRef] [PubMed]
  204. Salim, A.; Leman, J.A.; McColl, J.H.; Chapman, R.; Morton, C.A. Randomized comparison of photodynamic therapy with topical 5-fluorouracil in Bowen’s disease. Br. J. Dermatol. 2003, 148, 539–543. [Google Scholar] [CrossRef] [PubMed]
  205. Gross, K.; Kircik, L.; Ricorian, K.G. 5% 5-Fluorouracil cream for the treatment of small superficial basal cell carcinoma: Efficacy, tolerability, cosmetic outcome, and patient satisfaction. Dermatol. Surg. 2007, 33, 433–439. [Google Scholar] [CrossRef] [PubMed]
  206. Ishioka, P.; Maia, M.; Rodrigues, S.B.; Marta, A.C.; Hirata, S.H. Evaluation of the therapeutic results of actinic keratosis treated with topical 5% fluorouracil by reflectance confocal laser microscopy: Preliminary study. An. Bras Dermatol. 2015, 90, 426–429. [Google Scholar] [CrossRef] [PubMed]
  207. Kwon, S.; Dong, Z.M.; Wu, P.C. Sentinel lymph node biopsy for high-risk cutaneous squamous cell carcinoma: Clinical experience and review of literature. World J. Surg. Oncol. 2011, 9, 80. [Google Scholar] [CrossRef] [PubMed]
  208. Alam, M.; Ratner, D. Cutaneous squamous-cell carcinoma. N. Engl. J. Med. 2001, 344, 975–983. [Google Scholar] [CrossRef] [PubMed]
  209. Ross, A.S.; Schmults, C.D. Sentinel lymph node biopsy in cutaneous squamous cell carcinoma: A systematic review of the English literature. Dermatol. Surg. 2006, 32, 1309–1321. [Google Scholar] [CrossRef] [PubMed]
  210. Dona, E.; Veness, M.J.; Cakir, B.; Morgan, G.J. Metastatic cutaneous squamous cell carcinoma to the parotid: The role of surgery and adjuvant radiotherapy to achieve best outcome. ANZ J. Surg. 2003, 73, 692–696. [Google Scholar] [CrossRef] [PubMed]
  211. Reschly, M.J.; Messina, J.L.; Zaulyanov, L.L.; Cruse, W.; Fenske, N.A. Utility of sentinel lymphadenectomy in the management of patients with high-risk cutaneous squamous cell carcinoma. Dermatol. Surg. 2003, 29, 135–140. [Google Scholar] [PubMed]
  212. Martinez, J.C.; Cook, J.L. High-risk cutaneous squamous cell carcinoma without palpable lymphadenopathy: Is there a therapeutic role for elective neck dissection. Dermatol. Surg. 2007, 33, 410–420. [Google Scholar] [CrossRef] [PubMed]
  213. Garcia-Zuazaga, J.; Olbricht, S.M. Cutaneous squamous cell carcinoma. Adv. Dermatol. 2008, 24, 33–57. [Google Scholar] [CrossRef] [PubMed]
  214. Kyrgidis, A.; Tzellos, T.G.; Kechagias, N.; Patrikidou, A.; Xirou, P.; Kitikidou, K.; Bourlidou, E.; Vahtsevanos, K.; Antoniades, K. Cutaneous squamous cell carcinoma (SCC) of the head and neck: Risk factors of overall and recurrence-free survival. Eur. J. Cancer 2010, 46, 1563–1572. [Google Scholar] [CrossRef] [PubMed]
  215. Samsanavicius, D.; Kaikaris, V.; Norvydas, S.; Liubauskas, R.; Valiukeviciene, S.; Makstiene, J.; Maslauskas, K.; Rimdeika, R. Sentinel lymph node biopsy for high-risk cutaneous squamous cell carcinoma: Analysis of recurrence-free survival. Medicina 2016, 52, 276–282. [Google Scholar] [CrossRef] [PubMed]
  216. Durham, A.B.; Lowe, L.; Malloy, K.M.; McHugh, J.B.; Bradford, C.R.; Chubb, H.; Johnson, T.M.; McLean, S.A. Sentinel Lymph Node Biopsy for Cutaneous Squamous Cell Carcinoma on the Head and Neck. JAMA Otolaryngol. Head Neck Surg. 2016, 142, 1171–1176. [Google Scholar] [CrossRef] [PubMed]
  217. Ahadiat, O.; Higgins, S.; Sutton, A.; Ly, A.; Wysong, A. SLNB in cutaneous SCC: A review of the current state of literature and the direction for the future. J. Surg. Oncol. 2017, 116, 344–350. [Google Scholar] [CrossRef] [PubMed]
  218. Navarrete-Dechent, C.; Veness, M.; Droppelmann, N.; Uribe, P. High-risk cutaneous squamous cell carcinoma and the emerging role of sentinel lymph node biopsy: A literature review. J. Am. Acad. Dermatol. 2015, 73, 127–137. [Google Scholar] [CrossRef] [PubMed]
  219. Gore, S.; Shaw, D.; Martin, R.; Kelder, W.; Roth, K.; Uren, R.; Gao, K.; Davies, S.; Ashford, B.G.; Ngo, Q.; et al. Prospective study of sentinel node biopsy for high-risk cutaneous squamous cell carcinoma of the head and neck. Head Neck 2015, 38, E884–E889. [Google Scholar] [CrossRef] [PubMed]
  220. Maruyama, H.; Tanaka, R.; Fujisawa, Y.; Nakamura, Y.; Ito, S.; Fujimoto, M. Availability of sentinel lymph node biopsy for cutaneous squamous cell carcinoma. J. Dermatol. 2016, 44, 431–437. [Google Scholar] [CrossRef] [PubMed]
  221. Silberstein, E.; Sofrin, E.; Bogdanov-Berezovsky, A.; Nash, M.; Segal, N. Lymph Node Metastasis in Cutaneous Head and Neck Squamous Cell Carcinoma. Dermatol. Surg. 2015, 41, 1126–1129. [Google Scholar] [CrossRef] [PubMed]
  222. Martinez, J.; Cook, J. High-Risk Cutaneous Squamous Cell Carcinoma without Palpable Lymphadenopathy: Is There a Therapeutic Role for Elective Neck Dissection? Dermatol. Surg. 2007, 33, 410–420. [Google Scholar] [CrossRef] [PubMed]
Table 1. American Joint Committee on Cancer Tumor (T) Classification, 8th Edition [65].
Table 1. American Joint Committee on Cancer Tumor (T) Classification, 8th Edition [65].
SCC
TXPrimary tumor cannot be assessed
T0No evidence of primary tumor
TisCarcinoma in Situ
T1Tumor < 2 cm in greatest dimension
T2Tumor ≥ 2 cm and <4 cm in greatest dimension
T3Tumor ≥ 4 cm in greatest dimension and/or perineal invasion and/or deep invasion and/or minor bone erosion
T4aTumor with gross cortical bone/marrow invasion
T4bTumor with skull base invasion and/or skull base foramen involvement
Table 2. American Joint Committee on Cancer Tumor (N) Classification, 8th Edition [65].
Table 2. American Joint Committee on Cancer Tumor (N) Classification, 8th Edition [65].
SCC
Clinical N (cN)
NXRegional lymph nodes cannot be assessed
N0No regional lymph node metastasis
N1Metastasis in single ipsilateral lymph node, ≤3 cm in greatest dimension and ENE (−)
N2aMetastasis in single ipsilateral lymph node, >3 cm but not >6 cm in greatest dimension and ENE (−)
N2bMetastasis in multiple ipsilateral lymph node, none >6 cm in greatest dimension and ENE (−)
N2cMetastasis in bilateral or contralateral lymph nodes, none >6 cm in greatest dimension and ENE (−)
N3aMetastasis in a lymph node >6 cm in greatest dimension and ENE (−)
N3bMetastasis in any node (s) and clinically overt ENE (+)
Pathologic N (pN)
NXRegional lymph nodes cannot be assessed
N0No regional lymph node metastasis
N1Metastasis in single ipsilateral lymph node, ≤3 cm in greatest dimension and ENE (−)
N2aMetastasis in single ipsilateral lymph node, ≤3 cm in greatest dimension and ENE (+), or in single ipsilateral lymph node, >3 cm but not >6 cm in greatest dimension and ENE (−)
N2bMetastasis in multiple ipsilateral lymph nodes, none >6 cm in greatest dimension and ENE (−)
N2cMetastasis in bilateral or contralateral lymph, node (s), none >6 cm in greatest dimension and ENE (−)
N3aMetastasis in a lymph node >6 cm in greatest dimension and ENE (−)
N3bMetastasis in a single ipsilateral node >3 cm in greatest dimension and ENE (+); or multiple ipsilateral, contralateral, or bilateral nodes, any with ENE (+); or a single contralateral node ≤ 3 cm and ENE (+)
ENE–extranodal extension.
Table 3. American Joint Committee on Cancer Tumor (M) Classification, 8th Edition [65].
Table 3. American Joint Committee on Cancer Tumor (M) Classification, 8th Edition [65].
SCC
MXDistant metastasis cannot be assessed
M0No distant metastasis
M1Distant metastasis
Table 4. American Joint Committee on Cancer Tumor Staging, 8th Edition [65].
Table 4. American Joint Committee on Cancer Tumor Staging, 8th Edition [65].
SCC
StageTNM
0TisN0M0
IT1N0M0
IIT2N0M0
IIIT3N0 or N1M0
IVT1 or T2N1M0
T1, T2 or T3N2M0
Any TN3M0
T4Any NM0
Any TAny NM1
Table 5. National Comprehensive Cancer Network (NCCN) 2017 Guidelines for Risk Factors for Recurrence for basal cell carcinoma (BCC) and Recurrence and Metastasis for squamous cell carcinomas (SCC) [106].
Table 5. National Comprehensive Cancer Network (NCCN) 2017 Guidelines for Risk Factors for Recurrence for basal cell carcinoma (BCC) and Recurrence and Metastasis for squamous cell carcinomas (SCC) [106].
Low-RiskHigh-Risk
BCC
Location/sizeL < 20 mm
M < 10 mm
L > 20 mm
M ≥ 10 mm
H
BordersWell DefinedPoorly Defined
Primary vs. RecurrentPrimaryRecurrent
Immunosuppression(−)(+)
Site of Prior RT(−)(+)
Pathology
Subtype
Perineural Involvement
Nodular Superficial
(−)
Aggressive Growth Pattern
(+)
SCC
Location/sizeL < 20 mm
M < 10 mm
L > 20 mm
M ≥ 10 mm
H
BordersWell DefinedPoorly Defined
Primary vs. RecurrentPrimaryRecurrent
Immunosuppression(−)(+)
Site of Prior RT or Chronic Inflammatory Process(−)(+)
Rapidly Growing Tumor(−)(+)
Neurologic Symptoms(−)(+)
Pathology
Degree of Differentiation
Adenoid, Adenosquamous,
Desmoplastic, Metaplastic
Well or Moderately
Differentiated
(−)
Poorly Differentiated
(+)
Depth, Thickness or Clark Level<2 mm or I, II, III≥2 mm or IV, V
Perineural, Lymphatic, or Vascular Involvement(−)(+)
Table 6. Indications for Radiologic Imaging for Skin Cancers and Recommended Imaging Studies [107,108].
Table 6. Indications for Radiologic Imaging for Skin Cancers and Recommended Imaging Studies [107,108].
Possible Bony InvasionsCT *
Possible Orbit InvasionsCT-bony invasion, MRI **-soft tissue
Assessment the extent of Tumor Invasion in Soft TissueMRI
Staging of Lymph Nodes and Metastatic DiseaseCT or MRI or PET ***(PET-CT)
Evaluation for Potential Perineural SpreadMRI
Post-operative Surveillance for Recurrent DiseasePET-CT
* CT-computer tomography; ** MRI-magnetic resonance imaging; *** PET-positron emission tomography.

Share and Cite

MDPI and ACS Style

Fahradyan, A.; Howell, A.C.; Wolfswinkel, E.M.; Tsuha, M.; Sheth, P.; Wong, A.K. Updates on the Management of Non-Melanoma Skin Cancer (NMSC). Healthcare 2017, 5, 82. https://doi.org/10.3390/healthcare5040082

AMA Style

Fahradyan A, Howell AC, Wolfswinkel EM, Tsuha M, Sheth P, Wong AK. Updates on the Management of Non-Melanoma Skin Cancer (NMSC). Healthcare. 2017; 5(4):82. https://doi.org/10.3390/healthcare5040082

Chicago/Turabian Style

Fahradyan, Artur, Anna C. Howell, Erik M. Wolfswinkel, Michaela Tsuha, Parthiv Sheth, and Alex K. Wong. 2017. "Updates on the Management of Non-Melanoma Skin Cancer (NMSC)" Healthcare 5, no. 4: 82. https://doi.org/10.3390/healthcare5040082

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