Therapy Failure and Resistance Mechanism in Eyelid and Ocular Surface Tumors

 

Xiaojun Jua    Piotr Gacaa    Wanlin Fana    Alexander C. Rokohla,b    Yongwei Guoc    

Philomena A. Wawer Matosa,b    Steffen Emmertd    Vinodh Kakkasserye    

Ludwig M. Heindla,b

 

aDepartment of Ophthalmology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany, bCenter for Integrated Oncology (CIO) Aachen-Bonn-Cologne-Duesseldorf, Cologne, Germany, cEye Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China, dDepartment of Ophthalmology, University of Rostock, Rostock, Germany, eDepartment of Ophthalmology, University of Lübeck, Lübeck, Germany

 

 

 

 

Key Words

Periocular basal cell carcinoma • Squamous cell carcinoma • Sebaceous gland carcinoma • Conjunctival melanoma • Ocular surface squamous neoplasia • Therapeutics • Drug resistance

 

Abstract

Malignant tumors of the eyelids and ocular surface are common ocular malignancies. At present, surgical treatment is mostly the first choice for these types of tumors. However, postoperative tumor recurrence and metastasis are still regarded as failures in the treatment of such malignancies. Based on this, malignant tumors of the eyelid and ocular surface are sometimes accompanied by local adjuvant chemotherapy and systemic chemotherapy to treat patients with relapse, invasion of adjacent tissues, and systemic metastases. Still, drug resistance greatly affects the treatment effect. This review lists several mechanisms of recurrence and metastasis of ocular surface and eyelid tumors after surgery, as well as mechanisms that may lead to non-surgical treatment or drug resistance.

 

 

Introduction

 

Unlike most ophthalmic diseases, periocular and ocular malignancies affect vision and even threaten life. Managing periocular and ocular diseases, orbital or intracranial invasion, and metastatic lesions are challenging and often involve a multidisciplinary approach.

According to different epidemiological surveys, basal cell carcinoma (BCC) and squamous cell carcinoma (SCC) are the most common eyelid malignancies [1]. Sebaceous gland carcinoma (SGC) and Merkel cell carcinoma (MCC) are less common but more deadly, with relatively high rates of metastasis and death [1, 2]. Conjunctival melanoma (CM) and ocular surface squamous neoplasia (OSSN) are the most common conjunctival malignancies [3]. Among ocular malignancies, because tumors on the eyelids or ocular surface are more likely to receive early attention, malignant tumors in this area are usually treated by surgical resection after early diagnosis [4-8]. Recurrent or large tumors, orbital, intracranial or metastatic lesions often require alternative treatment options (combination of non-surgical methods, such as targeted therapies, chemotherapy and adjuvant radiotherapy [7, 9-12]. This review summarizes several mechanisms of recurrence and metastasis of the above eyelid and ocular surface tumors after surgery, as well as mechanisms that may lead to drug resistance to non-surgical treatment.

 

 

Postoperative recurrence and metastasis

 

Surgical excision is undeniably the most common treatment for eyelid and ocular tumors. Although most of them are not fatal, the incidence of recurrence and postoperative metastasis is usually regarded as a therapy failure. Table 1 [13-20] shows the recurrence and metastasis rates of the postoperative periocular malignant tumors. Excision with histologic margin assessment is the standard treatment modality for periocular tumors. After successful tumor excision, a histological examination should always be carried out to confirm the diagnosis and to check the excision margin to determine the resection status. The surgical resection should always excise as much malignant tissue as necessary to achieve an R0 resection. The safety distance is difficult to define for periocular malignancies since every millimeter of healthy tissue would be decisive for later functional reconstruction [21]. Thus, intraoperative margin control (IOMC) examines the tumor and its margins before reconstruction and is increasingly commonly used. It primarily involves Mohs micrographic surgery (MMS), fast-frozen controlled (FFC), and fast paraffin (FP), and predominantly affects the effect of surgical treatment and postoperative recurrence.

A postoperative histopathological examination is essential to confirm the diagnosis and determine the resection status to identify infiltrative growing subtypes; this also influences postoperative follow-up care and the prognosis [22, 23]. Aggressive tumor subtypes significantly affect the risk of a non-radical excision, leading to a high risk of postoperative recurrence [24]. Therefore, for any patient with periocular malignancy who underwent surgery, a regular, lifelong follow-up is necessary.

 

Table 1. The recurrence and metastasis rates of periocular malignant tumor after different surgical excision method

 

 

Failure and resistance of non-surgical treatment

 

Basal cell carcinoma

BCC is the most common eyelid malignancy worldwide, accounting for 90% of all cases [25]. It arises most frequently on the lower eyelid, followed by the inner canthus, upper eyelid and lateral canthus [26]. The tumor grows slowly and is mostly infiltrative and destructive [22, 27]. BCC is not fatal with rare distant metastases, local recurrence can be associated with about 20% of eyelid BCC cases [28]. Once metastasized, BCC is usually associated with an aggressive subtype of the primary tumor, and their overall prognosis tends to be worse than that of the primary one [29].

 

Hedgehog pathway inhibitor. The Hedgehog pathway is a pivotal event in the pathogenesis of BCCs, and it is reported to be activated by the PTCH1 gene, which is located on human chromosome 9q22 and encodes a transmembrane protein that negatively regulates smooth muscle (SMO), another transmembrane protein of the Hedgehog pathway [10, 30]. The mechanism of PTCH1 is binding to an extracellular ligand, such as the sonic hedgehog, then relieving the negative control on SMO, and SMO subsequently migrates in the cilia and activates the Gli transcription factor [31, 32]. Many of the genes regulated by Gli proteins are co-opted by cancer cells as they regulate several cancer-related processes, including proliferation, migration, invasion, and neovascularization [33-35].

If tumors have progressed deeply into tissues or distant metastasis, we call it locally advanced BCC (laBCC) or metastasis BCC (mBCC) [36]. In these cases, we can use Hedgehog pathway inhibitors as adjuvant therapy for patients who may suffer further disfigurement, functional loss and increased morbidity due to direct surgical treatment [36, 37].

Therefore, the occurrence of Hedgehog pathway mutations is considered a driving event in the formation of BCCs, and inactivation of the Hedgehog pathway has been a key therapeutic target for difficult-to-treat BCC [10]. Currently, two targeted hedgehog inhibitor therapies are available: Vismodegib and Sonidegib, which have different pharmacokinetics but target the same molecular SMO [38].

Vismodegib and Sonidegib play increasingly essential roles in adjuvant therapy. Still, some adverse events observed in prior studies, that include muscle spasms, dysgeusia, alopecia, fatigue, and weight loss, or serious such as fatal adverse events [35, 38]. Compared with Vismodegib, Sonidegib has the most serious and increasingly frequent side effects like CK (Creatine Kinase) elevation [34, 39]. These side effects are related to the pharmacological effects of SMO inhibitors. Even at low levels, treatment-related adverse events may lead to the therapy discontinuation in many patients [40]. Therefore, clinicians who can correctly predict and assess the treatment-related adverse events can improve patient compliance and develop treatment plans.

Another barrier in treating Hedgehog pathway inhibitors is resistance with progression and no response. It is described in a case series that regrowth of at least one tumor in 21% of patients with advanced BCCs after a mean of 56 weeks and confirmed that long-term efficacy is often mitigated by the development of tumor-acquired resistance to the medications [41]. Danial et al. showed that Sonidegib was less effective in Vismodegib-tolerant advanced BCC [42]. All patients demonstrated either progressive or stable disease with Sonidegib. The resistance mechanism is unclear, a murine model study suggests that it may be related to point mutations in SMO [35].

In the previous clinical observation of treatment of Vismodegib in advanced BCC, primary resistance occurred in 50% of patients, and secondary resistance occurred in 20% of patients [41, 43]. Primary drug resistance manifests as a mutation in SMO G497W, located in the most frequently mutated SMO region (exon 8e10) in BCC, and then a conformational rearrangement of the entire protein region, which eventually leads to a partial blockage of the protein drug entry site. This blockage leads to a significant decrease in the concentration of active Vismodegib at the SMO G497W binding site [44]. Secondary resistance showed mutations in PTCH1 and SMO D473Y after treatment. In these mutations, protein rearrangements occur, resulting in partial blockage of protein drug entry [44] (Fig. 1).

In addition, on-canonical HH pathway activation mediated by Gli, namely hedgehog- Gli (HH-GLI) signaling, also plays a key role in embryonic development, cell proliferation, differentiation and stem cell maintenance [45]. Amplification of Gli genes allows tumors to escape SMO inhibition, identity switching to more closely resembling stem cells of the isthmus, and the reduction of primary cilia, leading to a switch from the Hedgehog pathway to Ras/MAPK pathway [46]. It leads to insensitivity to conventional Hedgehog pathway inhibitors, resulting in drug resistance [47].

 

Fig. 1. Site of action and inhibit of different HPI on the hedgehog pathway. PTCH: patched: SMO: smoothened protein: SUFU: suppressor of the fused protein: GSK3B: glycogen synthetase kinase 3B; PKA: protein kinase A; GLI: transcription factor GLI; PDE: cyclin nucleotide phosphodiesterases; PI3K/Akt: phosphoinositide 3 kinase; HPI: hedgehog pathway inhibitors.

 

PD-L1. Programmed death ligand 1 (PD-L1) is a cell surface glycoprotein that is normally expressed by immune cells and binds to its receptor PD-1; it inhibits the immune response in the inflammatory response and promotes peripheral tolerance in normal physiological signaling. For many cancers, this pathway is exploited by the upregulation of PD-L1 on tumor cells and tumor-infiltrating lymphocytes (TIL), thereby evading immune surveillance [48].

Several laboratory and clinical studies suggest that PD-1 blockers should be considered salvage therapy for patients with advanced BCC whose disease has progressed after standard Hedgehog pathway inhibition [49].

The resistance mechanism to PD-1 therapy can be explained by immune escape [50]. Whereas HLA class I antigen down-regulation is widely recognized as a mechanism of tumor immune escape, both lack of HLA class I antigen expression and low numbers of activated cytotoxic T cells (CD8+/granzyme B+) can contribute to a lack of clinical response to PD-1 [51]. In BCC, HLA class I antigen down-regulation is associated with a deficiency of infiltrating CD8+ T cells [52]. In addition, β2-m expression, which plays a crucial role in HLA class I antigen expression, was not detected in BCC cells [53]. BCC tumors are infiltrated by more negative regulatory immune cells, which may also damage the activity of CD8+/granzyme B+ T cells and destroy the efficacy of PD-1 blocking [54].

 

Squamous cell carcinoma

Squamous cell carcinoma is the subsequent most common malignancy of the eyelid [55]. The tumor has a predilection for the lower eyelid and the lid margin. Clinically, SCC is indistinguishable from BCC, but it does not usually manifest surface vascularization and grows more rapidly. SCC most commonly occurs in fair-skinned elderly patients with a fair complexion and a history of chronic sun exposure and skin damage [10].

 

EGFR targeting for advanced periocular SCC. Epidermal Growth Factor Receptor (EGFR), is a transmembrane receptor protein in the ErbB receptor kinase family, also referred to as HER1 or ErbB1. EGFR is associated with various ligands, including EGF and transforming growth factor-α. Upon binding to these ligands, EGFR forms a homodimer or heterodimer with another member of the ErbB receptor family. Activation of EGFR in keratin-forming cells leads to induction or blockade of cell proliferation, differentiation, cell migration, and increased survival and resistance to apoptosis [56].

Many clinical studies have found that in metastatic SCC, EGFR is strongly expressed in all layers of skin in all cases compared to adjacent normal skin. At the same time, the degree of overexpression of EGFR is positively correlated with the aggressiveness of the disease [55, 57]. Furthermore, conjunctival SCC specimens showed moderate to strong EGFR expression in both invasive and in situ components [58].

Two EGFR inhibitors, Cetuximab and Erlotinib, are currently reported to be clinically effective in patients with locally advanced periocular SCC with orbital expansion [59]. Notably, 9-13% of SCC patients stop treatment due to the side effects of EGFR inhibitors [60]. Therefore, drug side effects such as skin toxicity, mucositis, esophagitis, corneal ulceration, conjunctivitis, and ectropion caused by EGFR inhibitors will be the focus of further research [61, 62]. Since the current literature records are less, it is still uncertain whether the resistance mechanism of EGFR inhibitors in periocular SCC is the same as that in other sites.

 

Sebaceous gland carcinoma

Sebaceous gland carcinoma (SGC) is a very rare and slowly growing tumor. It most frequently arises from the meibomian glands. It may also arise from the gland of Zeiss or sebaceous glands. The tumor occurs most frequently on the upper eyelid, where meibomian glands are more numerous [63]. Liang et al. reports that the incidence of SGC of the eyelid has significantly increased [64]. Compared to most other malignant eyelid tumors, its prognosis is still regarded as poor, and its mortality rate is second only to that of malignant melanoma [65]. The clinical diagnosis of SGC is frequently difficult because, external signs of malignancy may be subtle in its early stage, so the tumor may resemble a less aggressive lesion. In many cases, the delayed diagnosis of meibomian gland carcinoma is due to its ability to masquerade as various other ocular diseases such as chronic conjunctivitis, superior limbic keratoconjunctivitis, cicatricial pemphigoid chalazion, basal cell carcinoma, or other eyelid tumors [66]. Definitive diagnosis is only by pathology [67].

 

Topical mitomycin C chemotherapy. Pagetoid spread is a common feature of periocular sebaceous carcinoma, currently documented local chemotherapy for MMC (mitomycin C) is used in the treatment of SGC with pagetoid spread [68]. The most reason for MMC treatment disturb is short- and long- complications [69]. As for recurrence after treatment, the role of adjuvant MMC for SGC without pagetoid spread is not yet established in the literature. Furthermore, the effect of adjuvant MMC upon survival time and local recurrence in cases of SGC (with or without pagetoid spread) will need to be delineated through clinical trial. Given the infrequency of the diagnosis, such a trial would need to incorporate multiple centers over a considerable time period.

 

Neoadjuvant systemic chemotherapy. Although surgical excision is the primary treatment for localized SGCs, more advanced tumors may suggest topical therapy and neoadjuvant systemic chemotherapy. Because of the chemosensitivity of SGC, the choice of chemotherapy combination in eyelid SGC in various case reports has been extrapolated from the standard chemotherapy agents used in the management of head-and-neck squamous cell carcinoma, including platinum-based agents (cisplatin/ carboplatin) in combination with 5-fluorouracil and taxanes [70]. The current literature records, show few cases of neoadjuvant systemic chemotherapy used for SGC, and most of them are carried out in the form of adjuvant surgery. Therefore, further clinical observation and research are needed for study its drug resistance and failure.

 

Merkel cell carcinoma

Merkel cell carcinoma is a rare and aggressive neuroendocrine malignancy of the skin, which affects the elderly. Although MCC may occur in any skin area, 10% of tumors involve the eyelid or periocular area [71, 72]. This tumor commonly seen in the upper eyelid, especially near the eyelid margin, and usually results in partial or complete eyelash loss [12]. MCC presents as an asymptomatic, isolated nodule that may have an ulcerated surface or overlying dilated capillaries. MCC is a fast-growing tumor associated with sun exposure, immunosuppression, and polyomavirus [73]. Its rarity may lead to difficulty in diagnosis and delay in treatment of this highly malignant tumor. MCC is often misdiagnosed as a cyst, chalazion, keratoacanthoma, or BCC. It may be associated with concurrent tumors, such as SCC, BCC, SGC, breast and ovarian adenocarcinomas, lymphomas, and may share a carcinogenic process with other cells of neural crest origin [12, 74].

 

PD-L1. Until now, the PD-L1 inhibitor Avelumab is the only approved treatment for advanced MCC [75]. Avelumab is a human IgG1 monoclonal antibody, which binds to PD-L1 on cancer cells and blocks its interaction with PD-1 [76]. In the treatment of metastatic MCC, immune checkpoint blockade (ICB) shows a high response rate [76, 77]. Unfortunately, the immune checkpoint inhibitors (Avelumab) are only effective in a subset of patients. Patients who respond initially show a subsequent rapid disease progression due to primary and acquired resistance to PD-1/PD-L1 inhibition [78]. According to current reports, primary resistance seems to be a more important clinical problem in MCC [79]. However, follow-up studies are still few, so that the treatment resistance of MCC is still a pivotal point to be broken through.

Other targeted therapies should be the alternatives for patients with PD-1/PD-L1 treatment failure. Apoptosis inducers such as YM155, ABT-263 have been proved to induce cell death in MCC cell lines and xenograft models [80, 81]. Aberrant activation of the PI3K pathway may be a potential therapeutic target for Merkel cell carcinoma. Some clinical data and experimental results suggest that PI3K-mTOR pathway inhibition effectively treats MCC [82, 83]. A case report has reported that the selective PI3Kδ inhibitor Idelalisib is effective in metastatic MCC [84]. Inhibition of PI3Kδ interferes with B cell signaling and shifts the balance from immune tolerance to effective anti-tumor immunity by suppressing regulatory T cells and releasing cytotoxic T cells [85]. Therefore, targeted therapy has significant research prospects for treating MCC due to PD-1/PD-L1 treatment failure or resistance.

 

Conjunctival melanoma

Conjunctival melanoma may originate from primary acquired melanosis (PAM), nevi, or de novo. CM is a subtype of mucosal melanoma that arises from atypical melanocytes in the basal layer of the conjunctival epithelium and represents 2% of ocular tumors [86]. It is the second most frequent malignant neoplasm of the conjunctiva after SCC. Compared with uveal melanoma, the incidence of conjunctival melanoma is much lower [87]. They are most frequent in the interpalpebral region of the bulbar conjunctiva, they may be observed arising from the fornix, palpebral conjunctiva, and inner canthus.

 

MAPK pathway inhibitor. Mitogen-activated protein kinase (MAPK) pathways are evolutionarily conserved kinase modules that can link extracellular signals to mechanisms that control essential cellular processes including growth, proliferation, differentiation, migration, and apoptosis [88]. Many studies have evaluated the importance of the mitogen-activated protein kinase (MAPK) pathway in cutaneous melanoma [89]. Mutations common in cutaneous melanoma, such as V600E in BRAF exon 15, are also found in conjunctival melanoma [90]. Some studies found, BRAF mutations in 29% to 35% of conjunctival melanomas [91, 92]. Meanwhile, NRAS (neuroblastoma rat sarcoma viral oncogene) is also a critical mutation for tumor progression, and about 15% of melanomas contain NRAS mutations [93, 94]. Both NRAS or BRAF mutated oncogenes strongly activate the MAPK (MEK/ERK) pathway. Successful inhibitor therapy with specific BRAF and MEK inhibitors targeting the overactivated MAPK pathway driven by mutated NRAS or BRAF increases median overall survival. Therefore, specific inhibitors of these oncoproteins, MAPK pathway components or their combination have been used for tumor eradication. After a good initial response, resistant cells develop almost universally and greatly affect the drug’s effectiveness, causing treatment failure [88-90, 92, 93, 95].

 

Resistance to Mutated BRAF Inhibitors. Most melanomas show BRAF-V600E activating mutations. Currently there are BRAF inhibitory treatments, but many mechanisms are related to resistance to BRAF inhibitors [96]. BRAF-V600E activating mutations lead to constitutive activation of the MEK-ERK signaling pathway [97]. HGF/MET signaling is involved in resistance to BRAF inhibitors, HGF prevents cell death and restores activation of the MEK/ERK and PI3K-AKT signaling pathways by preventing cell death and restoring activation of oncogenes expressing BRAF-V600E [98, 99]. HGF expression analysis in BRAF-V600E melanoma patients is identified with its expression in the tumor-associated stroma. This expression increases after BRAFi treatment, which in turn increases resistance to BRAFi treatment [100].

POU Class 4 Homeobox 1 (POU Domain, Class 4, Transcription Factor 1, POU Class 4 Homeobox 1, POU4F1), a member of the POU domain family transcription factors, is also a stem cell-associated transcription factor expressed in proliferating precursor cells in the neural crest, POU4F1 may be involved in the resistance of melanoma to BRAFi. The resistance mechanism is that elevated POU4F1 activates the MEK/ERK pathway and thus the entire MAPK pathway, rendering melanoma resistant to BRAFi. POU4F1 directly binds to the promoters of MEK and MITF (Microphthalmia-associated Transcription Factor) to promote their expressions. POU4F1 is directly bound to the promoter region of MITF and transcriptionally promotes the expression of MITF, MITF contributes to the development of melanoma resistance to BRAFi [101] (Fig. 2).

Dual activation of the pathways greatly contributes to drug resistance, as these pathways promote cell survival and proliferation. Starting from the cell surface, several receptor tyrosine kinases (RTK) converge on parallel pathways, such as the MAPK and PI3K-Akt pathways. Upregulation of RTK has been shown to activate the MAPK pathway through RAS activation directly. In addition, upregulation of specific RTKs such as insulin-like growth factor 1 receptor (IGFR1) and platelet-derived growth factor receptor (PDGFR) can activate the PI3K-Akt pathway in a non-ERK-dependent manner. Epigenetic changes affecting the epidermal growth factor receptor (EGFR) have also been shown to induce the PI3K-Akt pathway in melanoma-resistant cells [102-106].

Recently, BRAF mutant melanoma cells have adapted to the feed-forward mechanism of MAPK targeted therapy by acquiring an auto-amplified CAF-like phenotype has been reported [108]. In addition to therapy-induced tumor secretomes, therapy-induced mechanical phenotypes could endow cancer cells with unique cell-autonomous abilities to survive and differentiate within challenging tumor-associated microenvironments, thereby contributing to drug resistance and relapse [107, 108].

 

Fig. 2. BRAF mutated melanoma responds to BRAF/MEK inhibitors-targeted therapy; Development of BRAF/MEK inhibitors-targeted therapy resistance. RTK: receptor tyrosine kinases; MITF: Melanocyte Inducing Transcription Factor; POU4F1: POU Domain, Class 4, Transcription Factor 1.

 

Resistance to Mutated NRAS Inhibitors. Neuroblastoma RAS viral oncogene homolog (NRAS) mutations exist in 15% to 20% of melanoma patients [105]. NRAS activates multiple effector pathways, and some mutant NRAS melanoma cells may be less dependent on MEK/ERK signaling pathway [109]. The major downstream effectors of RAS are RAF (a family of protein kinases, including A-RAF, B-RAF, C-RAF), phosphatidylinositol 3-kinase (PI3K), and Ral guanine exchange factor (RalGEF) [110]. Thus, PI3K and AKT activity may be important pathways in mutant NRAS melanoma cells resistant to MEK inhibitor treatment [111].

Romano et al. have recently revealed an E545K mutation in PIK3CA, the catalytic subunit of phosphatidylinositol 3 kinase PIK3 [112]. Unlike BRAF mutant melanoma, the activation of the MAPK pathway in NRAS mutant melanoma is achieved by activating the NRAS effector C-RAF instead of BRAF. It has been confirmed that S6 is the intersection between the MAPK and PI3K pathways. At the same time, S6 phosphorylation mainly depends on S6K1 [113]. Since PIK3CA-E545K increases S6K1 and S6 phosphorylation in the AKT/mTOR pathway, resulting in resistance to MEK inhibition [112].

CDK4/6 is another important drug inhibition point in NRAS mutant melanoma, increased CDK4/6 activity can lead to early G1-S cell cycle transition and promote the progression of melanoma [114]. RB is an oncosuppressor whose primary function, is to repress the transcription of genes required for the S-phase entry, preventing the unscheduled progression through the cell cycle. RB inhibits the transcription factors of the E2F family, which regulate genes involved in cell cycle control and mitotic progression [115]. After long-term treatment with CD4/6 inhibitors, cancer cells respond to inhibitory stress and re-enter the cell cycle. At the same time, the level of cyclin D1 rises. During the early adaptation period of CDK4/6i, the activated cyclin D1 takes effect. Thereby inactivating RB through phosphorylation, inducing transcriptional E2F activity, promoting the cell cycle re-entry after CDK4/6i treatment and causing drug treatment failure [116-118] (Fig. 3).

 

Fig. 3. NRAS mutated melanoma responds to inhibitors-targeted therapy.

 

Conjunctival intraepithelial neoplasia

Conjunctival intraepithelial neoplasia (CIN) includes a wide range of neoplastic intraepithelial changes ranging from dysplasia to full-thickness epithelial neoplasia or carcinoma in situ. Synonyms include mild, moderate and severe dysplasia, carcinoma in situ, ocular surface squamous neoplasia, intraepithelial epithelioma, and Bowenoid type of dyskeratosis [119].

Clinically, the lesions are sharply demarcated from surrounding normal epithelium and arise most typically at the corneal limbus, where corneal stem cells are located, with either or both conjunctival and corneal involvement. Most lesions are nonkeratinized, well-vascularized, pink, and have a raspberry-like configuration. The development is associated with ocular pigmentation, nonoffice and nonprofessional workers with a history of risk factors, such as sun exposure, fair skin, immunosuppression (organ transplant recipients), and smoking [4]. In addition, there may be a correlation between OSSN and co-infection with HPV types 6 and 11 (38%), HPV type 16 (30 to 58%), HPV type 18 (50-57%), HIV, or matrix metalloproteinase upregulation [120-124].

Although surgical resection is widely used in the treatment of OSSN, adjuvant treatments, including cryotherapy and local chemotherapy, are usually given after resection due to the high recurrence rate. The local chemotherapy drugs used for OSSN are mitomycin C (MMC), 5-fluorouracil (5-FU), anti-vascular endothelial growth factor (anti-VEGF), and interferon (IFN-α-2b) [125-127]. In recent years, antiviral drugs like cidofovir and Retinoic acid (RA) have also been used in the local treatment of OSSN [128, 129]. Currently, some of the ocular anticancer drugs used in OSSN are cytotoxic and neither specific nor targeted to cancer cells. Toxicity to healthy ocular tissues is inevitable. The ocular surface where OSSN is located is particularly susceptible to chemical toxicity because it consists of rapidly proliferating cell populations in the corneal epithelium, conjunctiva, skin and eyelid follicles. Based on the studies, the toxic effects of current topical antineoplastic drugs for OSSN are listed below (Table 2) [130, 131]. The hindrance of local chemotherapy comes from its side effects. Side effects caused by chemotherapeutic drugs often lead to treatment interruption, so treatment fails.

 

Table 2. Pharmacological and toxicological effects of different topical drugs used to treat OSSN

 

Adjuvant radiotherapy

Since excessive surgical excision may lead to anatomical dysfunction and poor aesthetic outcome, radiotherapy (RT) is an attractive treatment option for invasive tumors [132]. But for the side-effect of RT, has been recently reported inducing nonrepairable DNA damage and may stimulate the activation of immune system pathways [133]. Research has reported that over half of BCC patients induce various grades of acute or late toxicity after adjuvant radiotherapy [134]. By summarizing the recent literature on ocular radiotherapy, Table 3 [135-137] shows the adverse events of the RT in periocular tumors.

Recurrence after radiotherapy is still a therapy failure we should focus on. It reported overall recurrence at 5 years after the RT is 13.8% in SCC and 15.8% in BCC [138, 139]. In BCC patients, it is a strong correlation between subtype and gene expression in recurrence after radiotherapy, the sclerosing subtype and the high expression levels of p53 and Bcl-2 are the risk factor for recurrence [139]. Even in a recent single-center retrospective study, women were more likely than men to have disease progression after RT, but because of the relatively small number of patients, it remains to be seen whether gender is a real risk factor [140]. And in SCC patients, differentiation in SCCs affects the local recurrence rate [132]. For radiation treatment of conjunctival melanoma reported to date, tumor thickness and unfavorable tumor location (≥T2) are risk factors for local recurrence and metastatic disease; therefore, higher T-stage levels are associated with the risk of recurrence and metastatic disease [141]. Tumor origin is also a reported risk factor, there is a significantly higher risk for metastatic disease and death in conjunctival melanoma arising de novo [141]. Meanwhile, a retrospective study of 100 patients with eyelid sebaceous carcinoma showed that T categories (≥T2) and tumor size (>2 mm) were associated with an increased risk of local recurrence, lymph node metastasis, and distant metastasis after radiotherapy [142].

 

Table 3. Toxicological effects of radiotherapy in periocular malignancies

 

 

Conclusion

 

Considering that recurrent malignancies are more aggressive and express higher metastatic potential, patients with eyelid and ocular surface malignancies require postoperative regular tumor long-term follow-up. The frequency and duration of follow-up depend on the type of malignancy. A minimum of 5 years of follow-up is recommended for primary BCC or recurrent BCC of the scleroderma subtype, as well as SebCa, MCC, and SCC [5].

The complex mechanisms of recurrence and metastasis make it difficult to develop effective, safe and cost-effective therapies as well as convenient surveillance strategies for ocular malignancies. Given the recommendations in the regulation of uveal melanoma, we also advocate a data-driven surveillance program in eyelid and ocular surface malignancies with the ultimate goal of enrolling patients with oligometastatic and advanced disease in clinical trials to help develop effective regulatory and therapeutic approaches [143].

Meanwhile, to summarize the treatment of malignant eyelid and ocular surface tumors, single surgical treatment has a high recurrence rate. In contrast, recurrent malignant tumors are often associated with aggressive and multiple metastases. Local adjuvant therapy and systemic chemotherapy are essential. The side effects of local adjuvant therapy are also the reason that the current treatment interruption cannot be ignored. The response and outcome of systemic chemotherapy depends on multiple redundant and diverse biological processes and molecular mechanisms that affect the sensitivity of cancer cells to chemotherapeutic agents. There is a need to develop novel targeted cancer therapies that target cellular resistance, particularly "pivotal" genes. Understanding this multidimensional process of resistance mechanism in eyelid and ocular surface tumors, identifying and linking signaling cascades and regulatory genes involved, may pave the way for advanced molecular diagnostics, developing minimally invasive interventions, and the use of progenitor/stem cell and even regenerative therapy.

 

 

Acknowledgements

 

Author Contributions

(I) Conception and design: Xiaojun Ju, Ludwig M. Heindl; (II) Administrative support: Ludwig M. Heindl; (III) Provision of study materials: Ludwig M. Heindl, Alexander C. Rokohl, Xiaojun Ju; (IV) Collection and assembly of data: Alexander C. Rokohl, Xiaojun Ju, Wanlin Fan; (V) Data analysis and interpretation: Ludwig M. Heindl, Vinodh Kakkassery, Xiaojun Ju; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

 

Statement of Ethics

The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

 

 

Disclosure Statement

 

The authors have no conflicts of interest to declare.

 

 

References

 

1 Moran JM, Phelps PO: Periocular skin cancer: Diagnosis and management. Dis Mon 2020;66:101046.
https://doi.org/10.1016/j.disamonth.2020.101046

 

2 Zlatarova ZI, Dokova KG: Incidence of Non-melanoma Eyelid Malignancies in Bulgaria (2000-2015). Ophthalmic Epidemiol 2021;28:198-204.
https://doi.org/10.1080/09286586.2020.1808230

 

3 Shields CL, Chien JL, Surakiatchanukul T, Sioufi K, Lally SE, Shields JA: Conjunctival Tumors: Review of Clinical Features, Risks, Biomarkers, and Outcomes--The 2017 J. Donald M. Gass Lecture. Asia Pac J Ophthalmol (Phila) 2017;6:109-120.

 

4 Galor A, Karp CL, Oellers P, Kao AA, Abdelaziz A, Feuer W, Dubovy SR: Predictors of ocular surface squamous neoplasia recurrence after excisional surgery. Ophthalmology 2012;119:1974-1981.
https://doi.org/10.1016/j.ophtha.2012.04.022

 

5 Ho SF, Brown L, Bamford M, Sampath R, Burns J: 5 years review of periocular basal cell carcinoma and proposed follow-up protocol. Eye (Lond) 2013;27:78-83.
https://doi.org/10.1038/eye.2012.230

 

6 Luz FB, Ferron C, Cardoso GP: Surgical treatment of basal cell carcinoma: an algorithm based on the literature. An Bras Dermatol 2015;90:377-383.
https://doi.org/10.1590/abd1806-4841.20153304

 

7 Rock T, Bartz-Schmidt KU, Bramkamp M, Milla J, Bosmuller HC, Rock D: Clinical Management of Squamous Cell Carcinoma of the Conjunctiva. Am J Case Rep 2020;21:e919751.
https://doi.org/10.12659/AJCR.919751

 

8 Poignet B, Gardrat S, Dendale R, Lemaitre S, Lumbroso-Le Rouic L, Desjardins L, Cassoux N, Levy Gabriel C: Basal cell carcinomas of the eyelid: Results of an initial surgical management. J Fr Ophtalmol 2019;42:1094-1099.
https://doi.org/10.1016/j.jfo.2019.03.037

 

9 Venkateswaran N, Mercado C, Galor A, Karp CL: Comparison of Topical 5-Fluorouracil and Interferon Alfa-2b as Primary Treatment Modalities for Ocular Surface Squamous Neoplasia. Am J Ophthalmol 2019;199:216-222.
https://doi.org/10.1016/j.ajo.2018.11.007

 

10 Basset-Seguin N, Herms F: Update in the Management of Basal Cell Carcinoma. Acta Derm Venereol 2020;100:adv00140.
https://doi.org/10.2340/00015555-3495

 

11 Su MG, Potts LB, Tsai JH: Treatment of periocular basal cell carcinoma with neoadjuvant vismodegib. Am J Ophthalmol Case Rep 2020;19:100755.
https://doi.org/10.1016/j.ajoc.2020.100755

 

12 Yin VT, Merritt HA, Sniegowski M, Esmaeli B: Eyelid and ocular surface carcinoma: diagnosis and management. Clin Dermatol 2015;33:159-169.
https://doi.org/10.1016/j.clindermatol.2014.10.008

 

13 Sin CW, Barua A, Cook A: Recurrence rates of periocular basal cell carcinoma following Mohs micrographic surgery: a retrospective study. Int J Dermatol 2016;55:1044-1047.
https://doi.org/10.1111/ijd.13326

 

14 Weesie F, Naus NC, Vasilic D, Hollestein LM, van den Bos RR, Wakkee M: Recurrence of periocular basal cell carcinoma and squamous cell carcinoma after Mohs micrographic surgery: a retrospective cohort study. Br J Dermatol 2019;180:1176-1182.
https://doi.org/10.1111/bjd.17516

 

15 Mirzayev I, Gündüz AK, Özalp Ateş FS, Özcan G, Işık MU: Factors affecting recurrence after surgical treatment in cases with ocular surface squamous neoplasia. Int J Ophthalmol 2019;12:1426-1431.
https://doi.org/10.18240/ijo.2019.09.09

 

16 Harvey JA, Mirza SA, Erwin PJ, Chan AW, Murad MH, Brewer JD: Recurrence and mortality rates with different treatment approaches of Merkel cell carcinoma: a systematic review and meta-analysis. Int J Dermatol 2022;61:687-697.
https://doi.org/10.1111/ijd.15753

 

17 Singh S, Mohamed A, Kaliki S: Ocular surface squamous neoplasia: analysis based on the 8th American Joint Committee on Cancer classification. Int Ophthalmol 2019;39:1283-1291.
https://doi.org/10.1007/s10792-018-0943-x

 

18 Dudzisz-Sledz M, Sobczuk P, Kozak K, Switaj T, Kosela-Paterczyk H, Czarnecka AM, Falkowski S, Rogala P, Morysinski T, Spalek MJ, Zdzienicki M, Goryn T, Zietek M, Cybulska-Stopa B, Klek S, Kaminska-Winciorek G, Ziolkowska B, Szumera-Cieckiewicz A, Rutkowski P: Treatment of Locally Advanced Merkel Cell Carcinoma-A Multi-Center Study. Cancers (Basel) 2022;14:422.
https://doi.org/10.3390/cancers14020422

 

19 Litwin AS, Shah-Desai SD, Malhotra R: Two new cases of metastatic basal cell carcinoma from the eyelids. Orbit (Amsterdam, Netherlands) 2013;32:256-259.
https://doi.org/10.3109/01676830.2013.788668

 

20 Phan K, Oh LJ, Goyal S, Rutherford T, Yazdabadi A: Recurrence rates following surgical excision of periocular basal cell carcinomas: systematic review and meta-analysis. J Dermatolog Treat 2020;31:597-601.
https://doi.org/10.1080/09546634.2019.1698702

 

21 Kopecky A, Rokohl AC, Heindl LM: [Techniques for the Reconstruction of the Posterior Eyelid Lamella]. Klin Monbl Augenheilkd 2018;235:1415-1428.
https://doi.org/10.1055/a-0751-1069

 

22 Kakkassery V, Heindl LM: [SOP - Standarized procedures in diagnostics and therapies of periocular basal cell carcinoma]. Klin Monbl Augenheilkd 2017; DOI: 10.1055/s-0043-120086.
https://doi.org/10.1055/s-0043-120086

 

23 Hauschild A, Breuninger H, Kaufmann R, Kortmann RD, Klein M, Werner J, Reifenberger J, Dirschka T, Garbe C: Brief S2k guidelines--Basal cell carcinoma of the skin. J Dtsch Dermatol Ges 2013;11:10-15, 11-16.
https://doi.org/10.1111/ddg.12015_3

 

24 Gąsiorowski K, Iwulska K, Zapała J, Wyszyńska-Pawelec G: Periocular basal cell carcinoma: recurrence risk factors/when to reoperate? Postepy Dermatol Alergol 2020;37:927-931.
https://doi.org/10.5114/ada.2020.102109

 

25 Rogers HW, Weinstock MA, Feldman SR, Coldiron BM: Incidence Estimate of Nonmelanoma Skin Cancer (Keratinocyte Carcinomas) in the U.S. Population, 2012. JAMA Dermatol 2015;151:1081-1086.
https://doi.org/10.1001/jamadermatol.2015.1187

 

26 Walling HW, Fosko SW, Geraminejad PA, Whitaker DC, Arpey CJ: Aggressive basal cell carcinoma: Presentation, pathogenesis, and management. Cancer Metastasis Rev 2004;23:389-402.
https://doi.org/10.1023/B:CANC.0000031775.04618.30

 

27 Kakkassery V, Loeffler KU, Sand M, Koch KR, Lentzsch AM, Nick AC, Adamietz IA, Heindl LM: [Current diagnostics and therapy recommendations for ocular basal cell carcinoma]. Ophthalmologe 2017;114:224-236.
https://doi.org/10.1007/s00347-016-0370-1

 

28 Allali J, D'Hermies F, Renard G: Basal cell carcinomas of the eyelids. Ophthalmologica 2005;219:57-71.
https://doi.org/10.1159/000083263

 

29 Shi Y, Jia R, Fan X: Ocular basal cell carcinoma: a brief literature review of clinical diagnosis and treatment. Onco Targets Ther 2017;10:2483-2489.
https://doi.org/10.2147/OTT.S130371

 

30 Epstein EH: Basal cell carcinomas: attack of the hedgehog. Nat Rev Cancer 2008;8:743-754.
https://doi.org/10.1038/nrc2503

 

31 Briscoe J, Thérond PP: The mechanisms of Hedgehog signalling and its roles in development and disease. Nat Rev Mol Cell Biol 2013;14:416-429.
https://doi.org/10.1038/nrm3598

 

32 Hanna A, Shevde LA: Hedgehog signaling: modulation of cancer properies and tumor mircroenvironment. Mol Cancer 2016;15:24.
https://doi.org/10.1186/s12943-016-0509-3

 

33 Carballo GB, Honorato JR, de Lopes GPF, Spohr T: A highlight on Sonic hedgehog pathway. Cell Commun Signal 2018;16:11.
https://doi.org/10.1186/s12964-018-0220-7

 

34 Carpenter RL, Ray H: Safety and Tolerability of Sonic Hedgehog Pathway Inhibitors in Cancer. Drug Saf 2019;42:263-279.
https://doi.org/10.1007/s40264-018-0777-5

 

35 Leavitt E, Lask G, Martin S: Sonic Hedgehog Pathway Inhibition in the Treatment of Advanced Basal Cell Carcinoma. Curr Treat Options Oncol 2019;20:84.
https://doi.org/10.1007/s11864-019-0683-9

 

36 Silapunt S, Chen L, Migden MR: Hedgehog pathway inhibition in advanced basal cell carcinoma: latest evidence and clinical usefulness. Ther Adv Med Oncol 2016;8:375-382.
https://doi.org/10.1177/1758834016653605

 

37 Dreier J, Cheng PF, Bogdan Alleman I, Gugger A, Hafner J, Tschopp A, Goldinger SM, Levesque MP, Dummer R: Basal cell carcinomas in a tertiary referral centre: a systematic analysis. Br J Dermatol 2014;171:1066-1072.
https://doi.org/10.1111/bjd.13217

 

38 Dummer R, Ascierto PA, Basset-Seguin N, Dreno B, Garbe C, Gutzmer R, Hauschild A, Krattinger R, Lear JT, Malvehy J, Schadendorf D, Grob JJ: Sonidegib and vismodegib in the treatment of patients with locally advanced basal cell carcinoma: a joint expert opinion. J Eur Acad Dermatol Venereol 2020;34:1944-1956.
https://doi.org/10.1111/jdv.16230

 

39 Brancaccio G, Pea F, Moscarella E, Argenziano G: Sonidegib for the Treatment of Advanced Basal Cell Carcinoma. Front Oncol 2020;10:582866.
https://doi.org/10.3389/fonc.2020.582866

 

40 Basset-Séguin N, Hauschild A, Kunstfeld R, Grob J, Dréno B, Mortier L, Ascierto PA, Licitra L, Dutriaux C, Thomas L, Meyer N, Guillot B, Dummer R, Arenberger P, Fife K, Raimundo A, Dika E, Dimier N, Fittipaldo A, Xynos I, et al.: Vismodegib in patients with advanced basal cell carcinoma: Primary analysis of STEVIE, an international, open-label trial. Eur J Cancer 2017;86:334-348.
https://doi.org/10.1016/j.ejca.2017.08.022

 

41 Chang AL, Oro AE: Initial assessment of tumor regrowth after vismodegib in advanced Basal cell carcinoma. Arch Dermatol 2012;148:1324-1325.
https://doi.org/10.1001/archdermatol.2012.2354

 

42 Danial C, Sarin KY, Oro AE, Chang AL: An Investigator-Initiated Open-Label Trial of Sonidegib in Advanced Basal Cell Carcinoma Patients Resistant to Vismodegib. Clin Cancer Res 2016;22:1325-1329.
https://doi.org/10.1158/1078-0432.CCR-15-1588

 

43 Chang AL, Solomon JA, Hainsworth JD, Goldberg L, McKenna E, Day BM, Chen DM, Weiss GJ: Expanded access study of patients with advanced basal cell carcinoma treated with the Hedgehog pathway inhibitor, vismodegib. J Am Acad Dermatol 2014;70:60-69.
https://doi.org/10.1016/j.jaad.2013.09.012

 

44 Pricl S, Cortelazzi B, Dal Col V, Marson D, Laurini E, Fermeglia M, Licitra L, Pilotti S, Bossi P, Perrone F: Smoothened (SMO) receptor mutations dictate resistance to vismodegib in basal cell carcinoma. Mol Oncol 2015;9:389-397.
https://doi.org/10.1016/j.molonc.2014.09.003

 

45 Ingham PW, McMahon AP: Hedgehog signaling in animal development: paradigms and principles. Genes Dev 2001;15:3059-3087.
https://doi.org/10.1101/gad.938601

 

46 Kuonen F, Huskey NE, Shankar G, Jaju P, Whitson RJ, Rieger KE, Atwood SX, Sarin KY, Oro AE: Loss of Primary Cilia Drives Switching from Hedgehog to Ras/MAPK Pathway in Resistant Basal Cell Carcinoma. J Invest Dermatol 2019;139:1439-1448.
https://doi.org/10.1016/j.jid.2018.11.035

 

47 Pietrobono S, Gagliardi S, Stecca B: Non-canonical Hedgehog Signaling Pathway in Cancer: Activation of GLI Transcription Factors Beyond Smoothened. Frontiers in genetics 2019;10:556.
https://doi.org/10.3389/fgene.2019.00556

 

48 Chen J, Jiang CC, Jin L, Zhang XD: Regulation of PD-L1: a novel role of pro-survival signalling in cancer. Ann Oncol 2016;27:409-416.
https://doi.org/10.1093/annonc/mdv615

 

49 Lipson EJ, Lilo MT, Ogurtsova A, Esandrio J, Xu H, Brothers P, Schollenberger M, Sharfman WH, Taube JM: Basal cell carcinoma: PD-L1/PD-1 checkpoint expression and tumor regression after PD-1 blockade. J Immunother Cancer 2017;5:23.
https://doi.org/10.1186/s40425-017-0228-3

 

50 Sabbatino F, Marra A, Liguori L, Scognamiglio G, Fusciello C, Botti G, Ferrone S, Pepe S: Resistance to anti-PD-1-based immunotherapy in basal cell carcinoma: a case report and review of the literature. J Immunother Cancer 2018;6:126.
https://doi.org/10.1186/s40425-018-0439-2

 

51 Gettinger S, Choi J, Hastings K, Truini A, Datar I, Sowell R, Wurtz A, Dong W, Cai G, Melnick MA, Du VY, Schlessinger J, Goldberg SB, Chiang A, Sanmamed MF, Melero I, Agorreta J, Montuenga LM, Lifton R, Ferrone S, et al.: Impaired HLA Class I Antigen Processing and Presentation as a Mechanism of Acquired Resistance to Immune Checkpoint Inhibitors in Lung Cancer. Cancer Discov 2017;7:1420-1435.
https://doi.org/10.1158/2159-8290.CD-17-0593

 

52 Walter A, Barysch MJ, Behnke S, Dziunycz P, Schmid B, Ritter E, Gnjatic S, Kristiansen G, Moch H, Knuth A, Dummer R, van den Broek M: Cancer-testis antigens and immunosurveillance in human cutaneous squamous cell and basal cell carcinomas. Clin Cancer Res 2010;16:3562-3570.
https://doi.org/10.1158/1078-0432.CCR-09-3136

 

53 Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A: Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell 2017;168:707-723.
https://doi.org/10.1016/j.cell.2017.01.017

 

54 Sasidharan Nair V, Elkord E: Immune checkpoint inhibitors in cancer therapy: a focus on T-regulatory cells. Immunology and cell biology 2018;1:21-33.
https://doi.org/10.1111/imcb.1003

 

55 Maubec E, Duvillard P, Velasco V, Crickx B, Avril MF: Immunohistochemical analysis of EGFR and HER-2 in patients with metastatic squamous cell carcinoma of the skin. Anticancer Res 2005;25:1205-1210.

 

56 Yin VT, Pfeiffer ML, Esmaeli B: Targeted therapy for orbital and periocular basal cell carcinoma and squamous cell carcinoma. Ophthalmic Plast Reconstr Surg 2013;29:87-92.
https://doi.org/10.1097/IOP.0b013e3182831bf3

 

57 Ch'ng S, Low I, Ng D, Brasch H, Sullivan M, Davis P, Tan ST: Epidermal growth factor receptor: a novel biomarker for aggressive head and neck cutaneous squamous cell carcinoma. Hum Pathol 2008;39:344-349.
https://doi.org/10.1016/j.humpath.2007.07.004

 

58 Shepler TR, Prieto VG, Diba R, Neuhaus RW, Shore JW, Esmaeli B: Expression of the epidermal growth factor receptor in conjunctival squamous cell carcinoma. Ophthalmic Plast Reconstr Surg 2006;22:113-115.
https://doi.org/10.1097/01.iop.0000202609.92772.c3

 

59 El-Sawy T, Sabichi AL, Myers JN, Kies MS, William WN, Glisson BS, Lippman S, Esmaeli B: Epidermal growth factor receptor inhibitors for treatment of orbital squamous cell carcinoma. Arch Ophthalmol 2012;130:1608-1611.
https://doi.org/10.1001/archophthalmol.2012.2515

 

60 Heath CH, Deep NL, Nabell L, Carroll WR, Desmond R, Clemons L, Spencer S, Magnuson JS, Rosenthal EL: Phase 1 study of erlotinib plus radiation therapy in patients with advanced cutaneous squamous cell carcinoma. Int J Radiat Oncol Biol Phys 2013;85:1275-1281.
https://doi.org/10.1016/j.ijrobp.2012.09.030

 

61 Lewis CM, Glisson BS, Feng L, Wan F, Tang X, Wistuba, II, El-Naggar AK, Rosenthal DI, Chambers MS, Lustig RA, Weber RS: A phase II study of gefitinib for aggressive cutaneous squamous cell carcinoma of the head and neck. Clin Cancer Res 2012;18:1435-1446.
https://doi.org/10.1158/1078-0432.CCR-11-1951

 

62 Methvin AB, Gausas RE: Newly recognized ocular side effects of erlotinib. Ophthalmic Plast Reconstr Surg 2007;23:63-65.
https://doi.org/10.1097/IOP.0b013e31802d97f0

 

63 Dasgupta T, Wilson LD, Yu JB: A retrospective review of 1349 cases of sebaceous carcinoma. Cancer 2009;115:158-165.
https://doi.org/10.1002/cncr.23952

 

64 Kaliki S, Das A: Ocular and Periocular Tumors in India: An EyeSmart Electronic Medical Record Analysis of 9633 Cases from a Referral Center. 2020;27:199-203.
https://doi.org/10.4103/meajo.MEAJO_275_19

 

65 Rizvi SA, Maheshwari V: Advanced upper eyelid sebaceous gland carcinoma with deep orbital extension. Journal of surgical case reports 2010;1:5.
https://doi.org/10.1093/jscr/2010.1.5

 

66 Shields JA, Demirci H, Marr BP, Eagle RC, Jr., Shields CL: Sebaceous carcinoma of the eyelids: personal experience with 60 cases. Ophthalmology 2004;111:2151-2157.
https://doi.org/10.1016/j.ophtha.2004.07.031

 

67 Hurt MA, Weedon D: Weedon D. Weedon's Skin Pathology (2nd ed). London, United Kingdom, Churchill Livingstone, 2002.
https://doi.org/10.5826/dpc.0201a15

 

68 Margo CE: Periocular intraepithelial sebaceous neoplasia: critical appraisal of nomenclature and prognostic importance. J Clin Pathol 2020;73:172-175.
https://doi.org/10.1136/jclinpath-2019-206147

 

69 Russell HC, Chadha V, Lockington D, Kemp EG: Topical mitomycin C chemotherapy in the management of ocular surface neoplasia: a 10-year review of treatment outcomes and complications. Br J Ophthalmol 2010;94:1316-1321.
https://doi.org/10.1136/bjo.2009.176099

 

70 Verma S, Kumar N, Meel R, Sen S, Kashyap S, Sharma S, Pushker N, Pushpam D, Bakhshi S: Neoadjuvant Systemic Chemotherapy in Sebaceous Gland Carcinoma of the Eyelid: A Retrospective Study. Ocul Oncol Pathol 2021;7:251-256.
https://doi.org/10.1159/000515857

 

71 Xue Y, Thakuria M: Merkel Cell Carcinoma Review. Hematol Oncol Clin North Am 2019;33:39-52.
https://doi.org/10.1016/j.hoc.2018.08.002

 

72 Akhtar S, Oza KK, Wright J: Merkel cell carcinoma: report of 10 cases and review of the literature. J Am Acad Dermatol 2000;43:755-767.
https://doi.org/10.1067/mjd.2000.106505

 

73 Zwijnenburg EM, Lubeek SFK, Werner JEM, Amir AL, Weijs WLJ, Takes RP, Pegge SAH, van Herpen CML, Adema GJ, Kaanders J: Merkel Cell Carcinoma: New Trends. Cancers (Basel) 2021;13:1614.
https://doi.org/10.3390/cancers13071614

 

74 Vortmeyer AO, Merino MJ, Böni R, Liotta LA, Cavazzana A, Zhuang Z: Genetic changes associated with primary Merkel cell carcinoma. Am J Clin Pathol 1998;109:565-570.
https://doi.org/10.1093/ajcp/109.5.565

 

75 D'Angelo SP, Russell J, Lebbe C, Chmielowski B, Gambichler T, Grob JJ, Kiecker F, Rabinowits G, Terheyden P, Zwiener I, Bajars M, Hennessy M, Kaufman HL: Efficacy and Safety of First-line Avelumab Treatment in Patients With Stage IV Metastatic Merkel Cell Carcinoma: A Preplanned Interim Analysis of a Clinical Trial. JAMA Oncol 2018;4:e180077.
https://doi.org/10.1001/jamaoncol.2018.0077

 

76 Kaufman HL, Russell J, Hamid O, Bhatia S, Terheyden P, D'Angelo SP, Shih KC, Lebbé C, Linette GP, Milella M, Brownell I, Lewis KD, Lorch JH, Chin K, Mahnke L, von Heydebreck A, Cuillerot JM, Nghiem P: Avelumab in patients with chemotherapy-refractory metastatic Merkel cell carcinoma: a multicentre, single-group, open-label, phase 2 trial. Lancet Oncol 2016;17:1374-1385.
https://doi.org/10.1016/S1470-2045(16)30364-3

 

77 Nghiem PT, Bhatia S, Lipson EJ, Kudchadkar RR, Miller NJ, Annamalai L, Berry S, Chartash EK, Daud A, Fling SP, Friedlander PA, Kluger HM, Kohrt HE, Lundgren L, Margolin K, Mitchell A, Olencki T, Pardoll DM, Reddy SA, Shantha EM, et al.: PD-1 Blockade with Pembrolizumab in Advanced Merkel-Cell Carcinoma. N Engl J Med 2016;374:2542-2552.
https://doi.org/10.1056/NEJMoa1603702

 

78 Akinleye A, Rasool Z: Immune checkpoint inhibitors of PD-L1 as cancer therapeutics. J Hematol Oncol 2019;12:92.
https://doi.org/10.1186/s13045-019-0779-5

 

79 D'Angelo SP, Bhatia S, Brohl AS, Hamid O, Mehnert JM, Terheyden P, Shih KC, Brownell I, Lebbé C, Lewis KD, Linette GP, Milella M, Georges S, Shah P, Ellers-Lenz B, Bajars M, Güzel G, Nghiem PT: Avelumab in patients with previously treated metastatic Merkel cell carcinoma: long-term data and biomarker analyses from the single-arm phase 2 JAVELIN Merkel 200 trial. J Immunother Cancer 2020;8:e000674.
https://doi.org/10.1136/jitc-2020-000674

 

80 Dresang LR, Guastafierro A, Arora R, Normolle D, Chang Y, Moore PS: Response of Merkel cell polyomavirus-positive merkel cell carcinoma xenografts to a survivin inhibitor. PLoS One 2013;8:e80543.
https://doi.org/10.1371/journal.pone.0080543

 

81 Verhaegen ME, Mangelberger D, Weick JW, Vozheiko TD, Harms PW, Nash KT, Quintana E, Baciu P, Johnson TM, Bichakjian CK, Dlugosz AA: Merkel cell carcinoma dependence on bcl-2 family members for survival. J Invest Dermatol 2014;134:2241-2250.
https://doi.org/10.1038/jid.2014.138

 

82 Nardi V, Song Y, Santamaria-Barria JA, Cosper AK, Lam Q, Faber AC, Boland GM, Yeap BY, Bergethon K, Scialabba VL, Tsao H, Settleman J, Ryan DP, Borger DR, Bhan AK, Hoang MP, Iafrate AJ, Cusack JC, Engelman JA, Dias-Santagata D: Activation of PI3K signaling in Merkel cell carcinoma. Clin Cancer Res 2012;18:1227-1236.
https://doi.org/10.1158/1078-0432.CCR-11-2308

 

83 Kannan A, Lin Z, Shao Q, Zhao S, Fang B, Moreno MA, Vural E, Stack BC, Jr., Suen JY, Kannan K, Gao L: Dual mTOR inhibitor MLN0128 suppresses Merkel cell carcinoma (MCC) xenograft tumor growth. Oncotarget 2016;7:6576-6592.
https://doi.org/10.18632/oncotarget.5878

 

84 Shiver MB, Mahmoud F, Gao L: Response to Idelalisib in a Patient with Stage IV Merkel-Cell Carcinoma. N Engl J Med 2015;373:1580-1582.
https://doi.org/10.1056/NEJMc1507446

 

85 Ali K, Soond DR, Piñeiro R, Hagemann T, Pearce W, Lim EL, Bouabe H, Scudamore CL, Hancox T, Maecker H, Friedman L, Turner M, Okkenhaug K, Vanhaesebroeck B: Inactivation of PI(3)K p110δ breaks regulatory T-cell-mediated immune tolerance to cancer. Nature 2014;510:407-411.
https://doi.org/10.1038/nature13444

 

86 Shields CL, Shields JA: Ocular melanoma: relatively rare but requiring respect. Clin Dermatol 2009;27:122-133.
https://doi.org/10.1016/j.clindermatol.2008.09.010

 

87 McLaughlin CC, Wu XC, Jemal A, Martin HJ, Roche LM, Chen VW: Incidence of noncutaneous melanomas in the U.S. Cancer 2005;103:1000-1007.
https://doi.org/10.1002/cncr.20866

 

88 Dhillon AS, Hagan S, Rath O, Kolch W: MAP kinase signalling pathways in cancer. Oncogene 2007;26:3279-3290.
https://doi.org/10.1038/sj.onc.1210421

 

89 Wellbrock C, Arozarena I: The Complexity of the ERK/MAP-Kinase Pathway and the Treatment of Melanoma Skin Cancer. Front Cell Dev Biol 2016;4:33.
https://doi.org/10.3389/fcell.2016.00033

 

90 Gear H, Williams H, Kemp EG, Roberts F: BRAF mutations in conjunctival melanoma. Invest Ophthalmol Vis Sci 2004;45:2484-2488.
https://doi.org/10.1167/iovs.04-0093

 

91 Griewank KG, Westekemper H, Murali R, Mach M, Schilling B, Wiesner T, Schimming T, Livingstone E, Sucker A, Grabellus F, Metz C, Susskind D, Hillen U, Speicher MR, Woodman SE, Steuhl KP, Schadendorf D: Conjunctival melanomas harbor BRAF and NRAS mutations and copy number changes similar to cutaneous and mucosal melanomas. Clin Cancer Res 2013;19:3143-3152.
https://doi.org/10.1158/1078-0432.CCR-13-0163

 

92 Larsen AC, Dahl C, Dahmcke CM, Lade-Keller J, Siersma VD, Toft PB, Coupland SE, Prause JU, Guldberg P, Heegaard S: BRAF mutations in conjunctival melanoma: investigation of incidence, clinicopathological features, prognosis and paired premalignant lesions. Acta Ophthalmol 2016;94:463-470.
https://doi.org/10.1111/aos.13007

 

93 Raaijmakers MI, Widmer DS, Narechania A, Eichhoff O, Freiberger SN, Wenzina J, Cheng PF, Mihic-Probst D, Desalle R, Dummer R, Levesque MP: Co-existence of BRAF and NRAS driver mutations in the same melanoma cells results in heterogeneity of targeted therapy resistance. Oncotarget 2016;7:77163-77174.
https://doi.org/10.18632/oncotarget.12848

 

94 Griewank KG, Westekemper H, Murali R, Mach M, Schilling B, Wiesner T, Schimming T, Livingstone E, Sucker A, Grabellus F, Metz C, Süsskind D, Hillen U, Speicher MR, Woodman SE, Steuhl KP, Schadendorf D: Conjunctival melanomas harbor BRAF and NRAS mutations and copy number changes similar to cutaneous and mucosal melanomas. Clin Cancer Res 2013;19:3143-3152.
https://doi.org/10.1158/1078-0432.CCR-13-0163

 

95 Vachtenheim J, Ondrusova L: Many Distinct Ways Lead to Drug Resistance in BRAF- and NRAS-Mutated Melanomas. Life (Basel) 2021;11:424.
https://doi.org/10.3390/life11050424

 

96 Patel M, Eckburg A, Gantiwala S, Hart Z, Dein J, Lam K, Puri N: Resistance to Molecularly Targeted Therapies in Melanoma. Cancers (Basel) 2021;13:1115.
https://doi.org/10.3390/cancers13051115

 

97 Genomic Classification of Cutaneous Melanoma. Cell 2015;161:1681-1696.

 

98 Wilson TR, Fridlyand J, Yan Y, Penuel E, Burton L, Chan E, Peng J, Lin E, Wang Y, Sosman J, Ribas A, Li J, Moffat J, Sutherlin DP, Koeppen H, Merchant M, Neve R, Settleman J: Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors. Nature 2012;487:505-509.
https://doi.org/10.1038/nature11249

 

99 Fernandes M, Jamme P, Cortot AB, Kherrouche Z, Tulasne D: When the MET receptor kicks in to resist targeted therapies. Oncogene 2021;40:4061-4078.
https://doi.org/10.1038/s41388-021-01835-0

 

100 Straussman R, Morikawa T, Shee K, Barzily-Rokni M, Qian ZR, Du J, Davis A, Mongare MM, Gould J, Frederick DT, Cooper ZA, Chapman PB, Solit DB, Ribas A, Lo RS, Flaherty KT, Ogino S, Wargo JA, Golub TR: Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature 2012;487:500-504.
https://doi.org/10.1038/nature11183

 

101 Liu L, Yue Q, Ma J, Liu Y, Zhao T, Guo W, Zhu G, Guo S, Wang S, Gao T, Li C, Shi Q: POU4F1 promotes the resistance of melanoma to BRAF inhibitors through MEK/ERK pathway activation and MITF up-regulation. Cell Death Dis 2020;11:451.
https://doi.org/10.1038/s41419-020-2662-2

 

102 Villanueva J, Vultur A, Lee JT, Somasundaram R, Fukunaga-Kalabis M, Cipolla AK, Wubbenhorst B, Xu X, Gimotty PA, Kee D, Santiago-Walker AE, Letrero R, D'Andrea K, Pushparajan A, Hayden JE, Brown KD, Laquerre S, McArthur GA, Sosman JA, Nathanson KL, et al.: Acquired resistance to BRAF inhibitors mediated by a RAF kinase switch in melanoma can be overcome by cotargeting MEK and IGF-1R/PI3K. Cancer Cell 2010;18:683-695.
https://doi.org/10.1016/j.ccr.2010.11.023

 

103 Sullivan RJ, Flaherty KT: Resistance to BRAF-targeted therapy in melanoma. Eur J Cancer 2013;49:1297-1304.
https://doi.org/10.1016/j.ejca.2012.11.019

 

104 Wang J, Huang SK, Marzese DM, Hsu SC, Kawas NP, Chong KK, Long GV, Menzies AM, Scolyer RA, Izraely S, Sagi-Assif O, Witz IP, Hoon DSB: Epigenetic changes of EGFR have an important role in BRAF inhibitor-resistant cutaneous melanomas. J Invest Dermatol 2015;135:532-541.
https://doi.org/10.1038/jid.2014.418

 

105 Nazarian R, Shi H, Wang Q, Kong X, Koya RC, Lee H, Chen Z, Lee MK, Attar N, Sazegar H, Chodon T, Nelson SF, McArthur G, Sosman JA, Ribas A, Lo RS: Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation. Nature 2010;468:973-977.
https://doi.org/10.1038/nature09626

 

106 Sun C, Wang L, Huang S, Heynen GJ, Prahallad A, Robert C, Haanen J, Blank C, Wesseling J, Willems SM, Zecchin D, Hobor S, Bajpe PK, Lieftink C, Mateus C, Vagner S, Grernrum W, Hofland I, Schlicker A, Wessels LF, et al.: Reversible and adaptive resistance to BRAF(V600E) inhibition in melanoma. Nature 2014;508:118-122.
https://doi.org/10.1038/nature13121

 

107 Girard CA, Lecacheur M, Ben Jouira R, Berestjuk I, Diazzi S, Prod'homme V, Mallavialle A, Larbret F, Gesson M, Schaub S, Pisano S, Audebert S, Mari B, Gaggioli C, Leucci E, Marine JC, Deckert M, Tartare-Deckert S: A Feed-Forward Mechanosignaling Loop Confers Resistance to Therapies Targeting the MAPK Pathway in BRAF-Mutant Melanoma. Cancer Res 2020;80:1927-1941.
https://doi.org/10.1158/0008-5472.CAN-19-2914

 

108 Obenauf AC, Zou Y, Ji AL, Vanharanta S, Shu W, Shi H, Kong X, Bosenberg MC, Wiesner T, Rosen N, Lo RS, Massagué J: Therapy-induced tumour secretomes promote resistance and tumour progression. Nature 2015;520:368-372.
https://doi.org/10.1038/nature14336

 

109 Hodis E, Watson IR, Kryukov GV, Arold ST, Imielinski M, Theurillat JP, Nickerson E, Auclair D, Li L, Place C, Dicara D, Ramos AH, Lawrence MS, Cibulskis K, Sivachenko A, Voet D, Saksena G, Stransky N, Onofrio RC, Winckler W, et al.: A landscape of driver mutations in melanoma. Cell 2012;150:251-263.
https://doi.org/10.1016/j.cell.2012.06.024

 

110 Downward J: Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer 2003;3:11-22.
https://doi.org/10.1038/nrc969

 

111 Vu HL, Aplin AE: Targeting TBK1 inhibits migration and resistance to MEK inhibitors in mutant NRAS melanoma. Mol Cancer Res 2014;12:1509-1519.
https://doi.org/10.1158/1541-7786.MCR-14-0204

 

112 Romano G, Chen PL, Song P, McQuade JL, Liang RJ, Liu M, Roh W, Duose DY, Carapeto FCL, Li J, Teh JLF, Aplin AE, Chen M, Zhang J, Lazar AJ, Davies MA, Futreal PA, Amaria RN, Zhang DY, Wargo JA, et al.: A Preexisting Rare PIK3CA(E545K) Subpopulation Confers Clinical Resistance to MEK plus CDK4/6 Inhibition in NRAS Melanoma and Is Dependent on S6K1 Signaling. Cancer Discov 2018;8:556-567.
https://doi.org/10.1158/2159-8290.CD-17-0745

 

113 Gao MZ, Wang HB, Chen XL, Cao WT, Fu L, Li Y, Quan HT, Xie CY, Lou LG: Aberrant modulation of ribosomal protein S6 phosphorylation confers acquired resistance to MAPK pathway inhibitors in BRAF-mutant melanoma. Acta Pharmacol Sin 2019;40:268-278.
https://doi.org/10.1038/s41401-018-0020-z

 

114 Li A, Ma Y, Jin M, Mason S, Mort RL, Blyth K, Larue L, Sansom OJ, Machesky LM: Activated mutant NRas(Q61K) drives aberrant melanocyte signaling, survival, and invasiveness via a Rac1-dependent mechanism. J Invest Dermatol 2012;132:2610-2621.
https://doi.org/10.1038/jid.2012.186

 

115 Giacinti C, Giordano A: RB and cell cycle progression. Oncogene 2006;25:5220-5227.
https://doi.org/10.1038/sj.onc.1209615

 

116 Álvarez-Fernández M, Malumbres M: Mechanisms of Sensitivity and Resistance to CDK4/6 Inhibition. Cancer Cell 2020;37:514-529.
https://doi.org/10.1016/j.ccell.2020.03.010

 

117 Herrera-Abreu MT, Palafox M, Asghar U, Rivas MA, Cutts RJ, Garcia-Murillas I, Pearson A, Guzman M, Rodriguez O, Grueso J, Bellet M, Cortés J, Elliott R, Pancholi S, Baselga J, Dowsett M, Martin LA, Turner NC, Serra V: Early Adaptation and Acquired Resistance to CDK4/6 Inhibition in Estrogen Receptor-Positive Breast Cancer. Cancer Res 2016;76:2301-2313.
https://doi.org/10.1158/0008-5472.CAN-15-0728

 

118 Hayes TK, Luo F, Cohen O, Goodale AB, Lee Y, Pantel S, Bagul M, Piccioni F, Root DE, Garraway LA, Meyerson M, Johannessen CM: A Functional Landscape of Resistance to MEK1/2 and CDK4/6 Inhibition in NRAS-Mutant Melanoma. Cancer Res 2019;79:2352-2366.
https://doi.org/10.1158/0008-5472.CAN-18-2711

 

119 Kennerdell JS, Dekker A, Johnson BL, Dubois PJ: Fine-needle aspiration biopsy. Its use in orbital tumors. Arch Ophthalmol 1979;97:1315-1317.
https://doi.org/10.1001/archopht.1979.01020020057013

 

120 Di Girolamo N: Association of human papilloma virus with pterygia and ocular-surface squamous neoplasia. Eye (Lond) 2012;26:202-211.
https://doi.org/10.1038/eye.2011.312

 

121 Di Girolamo N, Atik A, McCluskey PJ, Wakefield D: Matrix metalloproteinases and their inhibitors in squamous cell carcinoma of the conjunctiva. Ocul Surf 2013;11:193-205.
https://doi.org/10.1016/j.jtos.2013.01.006

 

122 Watts P, Sullivan S, Davies S, Rao N, Stock D: Electromyography and computed tomography scan-guided fine needle aspiration biopsy of discrete extraocular muscle metastases. J AAPOS 2001;5:333-335.
https://doi.org/10.1067/mpa.2001.117572

 

123 Nassar DL, Raab SS, Silverman JF, Kennerdell JS, Sturgis CD: Fine-needle aspiration for the diagnosis of orbital hematolymphoid lesions. Diagn Cytopathol 2000;23:314-317.
https://doi.org/10.1002/1097-0339(200011)23:5<314::AID-DC5>3.0.CO;2-F

 

124 Sturgis CD, Silverman JF, Kennerdell JS, Raab SS: Fine-needle aspiration for the diagnosis of primary epithelial tumors of the lacrimal gland and ocular adnexa. Diagn Cytopathol 2001;24:86-89.
https://doi.org/10.1002/1097-0339(200102)24:2<86::AID-DC1016>3.0.CO;2-5

 

125 Frucht-Pery J, Rozenman Y, Pe'er J: Topical mitomycin-C for partially excised conjunctival squamous cell carcinoma. Ophthalmology 2002;109:548-552.
https://doi.org/10.1016/S0161-6420(01)00967-8

 

126 Karp CL, Galor A, Chhabra S, Barnes SD, Alfonso EC: Subconjunctival/perilesional recombinant interferon α2b for ocular surface squamous neoplasia: a 10-year review. Ophthalmology 2010;117:2241-2246.
https://doi.org/10.1016/j.ophtha.2010.03.052

 

127 Faramarzi A, Feizi S: Subconjunctival bevacizumab injection for ocular surface squamous neoplasia. Cornea 2013;32:998-1001.
https://doi.org/10.1097/ICO.0b013e318289ddd8

 

128 Krilis M, Tsang H, Coroneo M: Treatment of conjunctival and corneal epithelial neoplasia with retinoic acid and topical interferon alfa-2b: long-term follow-up. Ophthalmology 2012;119:1969-1973.
https://doi.org/10.1016/j.ophtha.2012.03.045

 

129 Ip MH, Robert George CR, Naing Z, Perlman EM, Rawlinson W, Coroneo MT: Topical Cidofovir for Treatment-Refractory Ocular Surface Squamous Neoplasia. Ophthalmology 2018;125:617-619.
https://doi.org/10.1016/j.ophtha.2017.11.021

 

130 Moon J, Choi SH, Lee MJ, Jo DH, Park UC, Yoon SO, Woo SJ, Oh JY: Ocular surface complications of local anticancer drugs for treatment of ocular tumors. Ocul Surf 2021;19:16-30.
https://doi.org/10.1016/j.jtos.2020.11.006

 

131 Pe'er J: Ocular surface squamous neoplasia: evidence for topical chemotherapy. Int Ophthalmol Clin 2015;55:9-21.
https://doi.org/10.1097/IIO.0000000000000050

 

132 Lazarevic D, Ramelyte E, Dummer R, Imhof L: Radiotherapy in Periocular Cutaneous Malignancies: A Retrospective Study. Dermatology 2019;235:234-239.
https://doi.org/10.1159/000496539

 

133 Surace L, Lysenko V, Fontana AO, Cecconi V, Janssen H, Bicvic A, Okoniewski M, Pruschy M, Dummer R, Neefjes J, Knuth A, Gupta A, van den Broek M: Complement is a central mediator of radiotherapy-induced tumor-specific immunity and clinical response. Immunity 2015;42:767-777.
https://doi.org/10.1016/j.immuni.2015.03.009

 

134 Kalaghchi B, Esmati E, Ghalehtaki R, Gomar M, Jaberi R, Gholami S, Babaloui S, Nabavi M, Sotoudeh S, Khanjani N, Kazemian A, Amouzegar-Hashemi F, Aghili M, Lashkari M: High-dose-rate brachytherapy in treatment of non-melanoma skin cancer of head and neck region: preliminary results of a prospective single institution study. J Contemp Brachytherapy 2018;10:115-122.
https://doi.org/10.5114/jcb.2018.75596

 

135 Thariat J, Martel A, Matet A, Loria O, Kodjikian L, Nguyen AM, Rosier L, Herault J, Nahon-Estève S, Mathis T: Non-Cancer Effects following Ionizing Irradiation Involving the Eye and Orbit. Cancers (Basel) 2022;14:1194.
https://doi.org/10.3390/cancers14051194

 

136 Jeganathan VS, Wirth A, MacManus MP: Ocular risks from orbital and periorbital radiation therapy: a critical review. Int J Radiat Oncol Biol Phys 2011;79:650-659.
https://doi.org/10.1016/j.ijrobp.2010.09.056

 

137 Krengli M, Masini L, Comoli AM, Negri E, Deantonio L, Filomeno A, Gambaro G: Interstitial brachytherapy for eyelid carcinoma. Outcome analysis in 60 patients. Strahlenther Onkol 2014;190:245-249.
https://doi.org/10.1007/s00066-013-0495-y

 

138 Barysch MJ, Eggmann N, Beyeler M, Panizzon RG, Seifert B, Dummer R: Long-term recurrence rate of large and difficult to treat cutaneous squamous cell carcinomas after superficial radiotherapy. Dermatology 2012;224:59-65.
https://doi.org/10.1159/000337027

 

139 Zagrodnik B, Kempf W, Seifert B, Müller B, Burg G, Urosevic M, Dummer R: Superficial radiotherapy for patients with basal cell carcinoma: recurrence rates, histologic subtypes, and expression of p53 and Bcl-2. Cancer 2003;98:2708-2714.
https://doi.org/10.1002/cncr.11798

 

140 Xu L, Tang X, Jiang N, Zhang S, Cao Y, Sun X: Radiation Therapy Efficacy and Toxicity for Orbital and Ocular Adnexal Mucosa-Associated Lymphoid Tissue (OAMALT): A Single-Center, Retrospective Study of 32 Cases. Cancer Manag Res 2021;13:8017-8024.
https://doi.org/10.2147/CMAR.S334396

 

141 Shields CL, Markowitz JS, Belinsky I, Schwartzstein H, George NS, Lally SE, Mashayekhi A, Shields JA: Conjunctival melanoma: outcomes based on tumor origin in 382 consecutive cases. Ophthalmology 2011;118:389-395.e1-2.
https://doi.org/10.1016/j.ophtha.2010.06.021

 

142 Sa HS, Rubin ML, Xu S, Ning J, Tetzlaff M, Sagiv O, Kandl TJ, Esmaeli B: Prognostic factors for local recurrence, metastasis and survival for sebaceous carcinoma of the eyelid: observations in 100 patients. Br J Ophthalmol 2019;103:980-984.
https://doi.org/10.1136/bjophthalmol-2018-312635

 

143 Tsai KK, Bollin KB, Patel SP: Obstacles to improving outcomes in the treatment of uveal melanoma. Cancer 2018;124:2693-2703.
https://doi.org/10.1002/cncr.31284