Abstract
One in twenty solid organ transplant recipients (SOTRs) will develop a highly morbid or fatal cutaneous carcinoma after transplantation. The majority of these cases develop on the head and neck and may require intervention on the part of dermatology, dermatologic surgery, otolaryngology, transplant medicine, radiation oncology, and medical oncology. In this review, we discuss the problem of cutaneous squamous cell carcinoma (cSCC) in SOTRs as well as the prognostic factors and management strategies to care for this population.
Keywords: cutaneous head and neck cancer, transplant, squamous cell carcinoma, immunosuppression, immunotherapy
INTRODUCTION
Cutaneous squamous cell carcinoma (cSCC) is a malignancy of keratinocytes and is the second most common cancer with an annual incidence of 700,000 cases resulting in nearly 8800 deaths in the United States each year[1,2]. Unfortunately, the incidence of cSCC has continued to rise with an increase of 50 to 200% over the last 3 decades[1,3]. Given the role of the immune system in surveillance and removal of dysplasia, it is not surprising that immunosuppression leads to an increase in incidence of cSCC. Solid organ transplant recipients (SOTRs) have a 65–250 times increased incidence of cSCC and the risk is correlated with increased doses of immunosuppression, with increased risk with higher levels of immunosuppression, such as in lung transplant recipients [4–8]. Beyond just an increase in incidence, cSCC in SOTRs tends to recur more often and behave more aggressively with increased rates of metastasis and death. [9–11] The skin cancer specific mortality for transplant patients is nine-fold higher than those arising in immunocompetent patients. [12] Moreover, one in twenty SOTRs have a highly-morbid or fatal cSCC after transplant[13]. Therefore, there is a need for improvements in prevention, detection, and management of this disease in this patient population. In this review, we summarize the current data on keratinocyte carcinogenesis in SOTRs, review prognostic factors, and define the management of cSCC in SOTRs.
KERATINOCYTE CARCINOGENESIS IN SOTRs
The majority of cSCC occurs in sun-exposed locations and is driven by ultraviolet radiation (UVR) from sunlight and tanning beds. UVR functions as a carcinogen via two pathways leading to DNA mutagenesis: 1. direct DNA photoproduct formation and interaction with intracellular photosensitizers leading to the generation of reactive oxygen species and 2. Downregulation of the local immune response in the skin by decreasing antigen presentation capacity and increasing tolerance (reviewed in [14]). In SOTRs, immune surveillance is globally suppressed by drug therapy directed at T cells in an effort to prevent rejection. Therefore, the local environment is permissive for perpetuation of dysplasia and development of invasive carcinomas.
Chronic UVR exposure leads to mutations in known tumor suppressor genes (TP53, NOTCH1, FAT1) and known oncogenes (HRAS, KRAS, NRAS). In fact, the mutation rate in sun-exposed but otherwise normal appearing skin is estimated to be 5 mutations per megabase based on studies of upper eyelid skin which is close to the estimated mutational burden of oropharyngeal squamous cell carcinomas but about 10 fold lower than that reported for cSCCs [15]. Genome sequencing of cSCC has revealed nearly universal inactivating or non-synonymous mutations in TP53 [16].
More recent work suggests that the immunosuppressive drugs given to SOTRs may also contribute to specific mutations leading to oncogenesis. Work by Inman and colleagues demonstrated a gene mutation signature seen in cSCC arising in patients on azathioprine [16]. The prevalence of this signature correlated with the time the patient had been on the drug. Interestingly, this gene signature was distinct from those attributed to UVR and was heavily biased to the transcriptional strand, therefore leading the authors to hypothesize that it may influence transcription coupled nucleotide excision repair. In cell culture experiments, tacrolimus and mycophenolate were also shown to impair nucleotide excision repair pathways which are often used by keratinocytes to combat UVR-induced base substitution [17]
Furthermore, increasing evidence suggests that the drugs given to modulate T cell immunity may have a carcinogenic effect by modulation of transcription factors. Calcineurin inhibitors, the backbone of many immunosuppression regimens, can modulate ATF3 in keratinocytes leading to unchecked proliferation [18]. Therefore, these drugs also appear to be playing a role in cellular function in cells not involved in immunity. Continued investigation of the molecular changes of malignant keratinocytes from cSCC arising in the immunosuppressed patient is warranted.
Voriconazole, the triazole antifungal, used to prevent Aspergillus infection after lung transplant, has been implicated in phototoxic drug reactions and as an agent leading to increase in cSCC[19]. In a retrospective cohort study encompassing 20 years of lung transplantation at a single center, there was a 73% increase in the risk of cSCC in patients who received voriconazole [20]. The mechanism for voriconazole genotoxicity is likely due to increased oxidative damage[21,22].
Unlike oropharyngeal squamous cell carcinoma where HPV has a defined role in oncogenesis, there is conflicting data on the role of human papilloma virus (HPV) in cSCC carcinogenesis with the exception of lesions of the genitalia [23]. The skin has the highest prevalence of HPV when compared to other organ systems, however the subtypes of alpha HPVs found in the skin tend to be of low-oncogenic risk and often lead to verruca vulgaris (common wart) formation. Members of the skin-tropic beta genus of HPV which rarely integrate in the host genome have been found in verruca plana (flat warts) and are hypothesized to cause progression to carcinoma as patients with epidermodysplasia verruciformis are particularly susceptible to HPV beta infection and develop cSCCs early in adulthood. With an abundance of subtypes of beta HPV, high rate of colonization, and no clear mechanism of carcinogenesis it remains unclear what role beta HPVs cause in cSCC. A recent paper implicates CD8 T cell response to beta HPVs in protection from carcinogenesis and notes loss of that response in immunosuppressed individuals[24]. Unlike oropharyngeal squamous cell carcinoma, p16 is not a reliable marker for HPV etiology in cSCC as 100% of the invasive cSCCs tested showed staining for p16 in a retrospective analysis [25]. Therefore, the role of HPV in cSCC carcinogenesis remains unclear in immunosuppressed patients.
PROGNOSTIC FACTORS
Staging
The expert consensus-based American Joint Committee on Cancer staging system 7th edition (AJCC7) did not optimally stratify patients, as patients with poor outcomes were enriched in stage T2, whereas few patients were classified as T3 or T4 [1]. A study in heart and lung transplant patients showed an increase in risk of recurrence with increasing stage when using the AJCC7 [26] however no patients met criteria for T3 or T4 in this cohort. Subsequently, the Brigham and Women’s Hospital Tumor (BWT) Staging of cSCC was introduced to better stratify the T2 of the AJCC7 [27]. Out of the 84 patients upstaged from T2 in AJCC7 to T2b in BWT, 18 (21%) had a local recurrence, 18 (21%) a nodal metastasis and 9 (11%) disease specific death [28], reflecting the high-risk nature of T2b by BWT criteria. Consequently, the AJCC 8th edition aimed to shift higher risk patients from T2 to T3 [29], while merging the highest risk patients from the former AJCC7 T3 and T4 stages in a new T4a and T4b stage. The clinical and pathological factors used for tumor staging are summarized in Table 1. A recent study has confirmed, that the AJCC8 is more distinctive, monotonous as well as homogeneous than the AJCC7, while being comparable to the BWT [30]. Blechman and colleagues demonstrated similar distinctiveness, homogeneity, and monotonicity in AJCC8 and BWH staging when assessing cSCCs from 58 immunosuppressed patients [31]
Table 1.
Staging System | Stage | High Risk Clinical Features | High Risk Pathological Features |
---|---|---|---|
AJCC (8th Edition) | Tis= in situ disease T1=no high risk features T2=moderate size T3=large size or one high risk pathological feature T4a=cortical bone/marrow invasion T4b=skull base invasion and/or skull base foramen involvement |
Moderate size (2–4cm) Large size >4cm |
Perineural invasion (nerves deeper than dermis, >=0.1mm in caliber, or with radiological or clinical evidence of involvement) Deep Invasion (beyond subcutaneous fat or >6mm from granular layer) |
BWH | Tis= in situ disease T1= no high risk features T2a= one high risk feature T2b= 2–3 high risk features T3= 4 high risk features or bone involvement |
Size >= 2cm | Poor differentiation PNI of nerves >= 0.1mm in caliber Invasion beyond subcutaneous fat |
A comprehensive meta-analysis of 17,248 patients has validated the relevance for many of the AJCC8 factors in predicting local recurrence, metastasis or disease-specific death, yet some criteria shown to be independently and strongly correlated with outcome measures, such as immunosuppression and location (lip, temple, ear), are not included in either the BWH or the AJCC8 system [2]. This observation suggests that immunosuppression may lead to biologically aggressive cancer behavior that is not currently captured by pathologic staging factors (perineural invasion, depth of invasion, grade of differentiation). An obstacle to including immunosuppression as a factor in staging is the heterogeneity of suppression, both in terms of underlying cause of immunosuppression (organ transplantation, HIV infection, chronic lymphocytic leukemia) and treatment (specific drug, dose, duration of treatment) [2,29]. We recommend considering all invasive cSCCs with a high-risk pathologic feature in SOTRs to be at elevated risk for recurrence and metastasis and therefore advocate for an intensified workup, treatment, and surveillance.
MANAGEMENT
Screening
Due to the increased risk of cSCCs in SOTRs, clinical skin surveillance is an integral part of management of these patients. Patients with Fitzpatrick skin phototype I (always burn with exposure to sunlight) and II (often burn, rarely tan with exposure to sunlight) are at the highest risk for cSCC development. Further, patients who are male, over 50 at the time of the transplant, and have a heart or lung transplant are at the highest risk of cSCC [8]. Currently, there is no validated risk stratification system available for stratifying patients pre- or post-transplant. Rather, an expert consensus panel has formulated recommendations for screening in the post-transplant setting. Patients without lesions should have skin screening every 12 months while patients with one skin cancer should have skin exam every 3–6 months. In patients with multiple nonmelanoma skin cancers or lesions at high risk for recurrence or metastasis, screening should be done every 3 months [32].
Primary and Secondary Prevention
As UV light is the major driver of cSCC, UV protection is of utmost importance for prevention of cancer in this population. All fair-skinned transplant recipients should be counseled on sunscreen use, sun protective clothing, and avoidance of midday sun. Numerous studies have looked at sun safety education for solid organ transplant recipients. Collectively, these studies have shown improvement in patient understanding and compliance with sun protection with all modalities of teaching [33,34]. A study on sunscreen use and malignancy revealed fewer precancers as well as cSCC in SOTRs who used sunscreen on a regular basis [35].
Taking advantage of the march to carcinogenesis which starts as normal skin progresses to actinic keratosis (AK), followed by cSCC in situ, and finally invasive cSCC, patients can be treated with field therapy for field disease. Field disease is characterized by a large plaque of AK in a sun exposed area and typically has ill-defined borders[36]. Given the pathophysiology of carcinogenesis, it is not surprising that these patients would have significant field disease burden as adjacent skin is typically exposed to the same amount of UV radiation. Topical therapies to ameliorate field cancerization have been recommended. The most common of these is topical 5-fluorouracil. This drug blocks DNA synthesis and has shown improvement in field disease in SOTRs with 98% AK clearance rate at 8 weeks post therapy [37,38]. Capecitabine, the oral pro-drug for 5-fluorouracil has been used with some success for treatment of field disease and secondary prevention. In two case observation studies of SOTRs, a decrease from one cSCC every two months, to one every six months was seen when the patient was placed on capecitabine at 1g/m2 divided into two daily doses, given for 14 days followed by a 7 day drug holiday. [39,40]. However, in both reports, 60% of patients required a break from the medication regimen due to toxicity. Additionally, imiquimod, a Toll-like receptor agonist, has been used in transplant recipients to illicit a local immune response to clear actinic damage. Initially there were concerns of inciting a systemic immune response with use of this medication, however two RCTs in renal transplant recipients showed efficacy without adverse effect to the graft [41,42]. One treatment session lead to 49% AK clearance in SOTRs at one month post treatment [43].
Photodynamic Therapy (PDT) utilizes the photosensitizing products of the heme pathway to cause cell death after application of wavelength restricted visible light. Recent work has shown that this treatment can normalize aberrant cancer-associated gene expression pathways [44]. PDT was found to be more effective at clearing field disease than topical chemotherapy [37] and imiquimod [43] in SOTRs. Some studies have suggested primary and secondary prevention of both field disease and cSCC with PDT for SOTRs, whereas others have not shown significant benefit [43,45–48].
A commonly prescribed medication in the dermatology clinic is the systemic retinoid, acitretin. While topical retinoid (tretinoin cream) was unable to improve field disease and prevent cSCC development in the Veteran’s Study, systemic retinoid has been used successfully to slow down development of cSCC in the high risk population [49,50]. However, many report side effects from the medication and a rebound effect when the medication is removed. With known teratogenicity, this medication should not be used in women of child-bearing potential. More recently, work in Australia suggested the addition of niacinamide to the diet (500mg twice a day) for secondary prevention of skin cancer in all-comers [51]. However, a study of nicotinamide in patients who had undergone renal transplant did not show an improvement, perhaps due to the study being underpowered due to lack of enrollment [52].
Modification of Immunosuppression Regimen
Expert consensus guidelines suggest reducing immunosuppression in transplant patients who develop multiple skin cancers per year and in individuals with high risk lesions [53]. For patients on an immunosuppression regimen containing a cell cycle inhibitor, such as azathioprine or mycophenolate, removal of the cell cycle inhibitor is paramount. A meta-analysis revealed a 56% increase in risk for cSCC in patients exposed to azathioprine [54].
Previous studies have shown that sirolimus, a macrolide that inhibits mTOR therefore blocking central cell growth and proliferation signaling pathways, has anti-tumor properties, particularly in patients with renal transplants with history of cSCC [55,56]. A switch from tacrolimus to sirolimus was found to prevent cSCC development, if done after the patient has one cSCC, regardless of whether that one tumor was considered high risk [56]. The addition of sirolimus lead to a reduction in the incidence of skin cancer and lower risk of recurrence with no increased risk of overall mortality [57]. Although adverse effects are common in patients on sirolimus, serious consideration should be given to transitioning transplant patients with cSCC patients off calcineurin inhibitors which have been associated with increased incidence of cSCC and recurrent lesions [58].
Surgical Resection
Local recurrence and nodal metastasis are the main determinants of morbidity and mortality both in the immunocompetent and immunosuppressed population, therefore locoregional control is of paramount importance. Surgical management of advanced cSCC in the head and neck region in immunocompromised patients can be challenging. Surgeons must balance extent of surgery (margins, regional nodal dissection) with potential cosmetic and functional morbidity. Tumors often encroach upon or involve key facial and cervical structures including the eye, nose, lips, and ears. Moreover, the primary tumor or pathologic regional disease may invade into deeper structures such as facial muscles, parotid gland, bone, and facial nerve. Surgical excision techniques which allow for complete circumferential peripheral and deep margin assessment (CCPDMA) should be utilized in immunosuppressed patients.
Mohs micrographic surgery provides complete margin assessment. However, when Mohs surgery is unavailable or unfeasible, traditional excision with CCPDMA by frozen sections in place of the breadloaf technique is an alternative [59–61]. A combined procedure where peripheral margins are cleared by a Mohs surgeon and deep margin is cleared by an otolaryngologist, surgical oncologist, or plastic surgeon, may also be considered for more extensive cases. Additionally, a staged-procedure with en face grossing, fixing, and paraffin embedding of the sample such that all margins are examined to ensure complete margin assessment may suffice. In many cases, surgical resection is necessary to complete tumor staging. In a recent report from a single institution, over 70% of T3 tumors were only appropriately staged after information gained from pathologic analysis during Mohs surgery [62]. Therefore, surgeons should consider sending the debulked section of the tumor for fixation and histopathologic analysis for appropriate staging. There is encouraging evidence that the complete margin assessment offered by Mohs or CCPDMA indeed ensures local control with recurrence rates between 1–5% in the immunocompetent patient [63,64]. In a single center cohort of 215 patients, 20% of which were immunosuppressed, the recurrence rates of high-risk cSCC after Mohs surgery was 1.5% [65]. The immunosuppressed patients were not individually analyzed from this cohort.
Though it is well known that there is increased risk of regional metastases in the immunocompromised patient (5–12%) with cSCC, there is no clear consensus for surgical management of regional nodal basins in the clinically node-negative patient[66]. In this situation, surgeons often consider regional basin dissection when there are multiple risk factors including advanced primary disease(>2 cm in size), deep tumor invasion (beyond subcutaneous fat, bone invasion), concerning histologic features (poorly differentiated, perineural invasion), or in cSCC involving high-risk areas such as the external ear or lip [67]. Alternatively, high risk patients have also been stratified by AJCC 8th edition staging and Brigham and Women’s Tumor staging system. Interrogating the sentinel node might be especially useful in the AJCC8 T3 stage where 30% of patients with poor histological differentiation show lymph node metastasis [30]. A meta-analysis of 23 studies showed, that 7.9% of patients present with a positive sentinel lymph node (SLN), while at the same time acknowledging that the underlying primary studies might have been limited by a unclear definition of high risk patients by the respective staging systems [68]. More data, utilizing an updated staging system, is needed to show the clinical benefit of sentinel lymph node biopsies in the high-risk population. We recommend close clinical and sonographic surveillance of the regional lymph node basin for all immunosuppressed patients until the benefit of SLN biopsy has been shown more clearly [69]. Future investigations may reveal which patients would be best suited for observation, SLNB, or elective regional basin dissection in immunocompromised patients.
When regional metastatic disease is present, surgical excision via neck dissection and/or parotidectomy remain the standard of care. When parotid metastases are present, incidence of occult disease in the neck is high (22.5– 42%) in all patients with head and neck cSCC [70,71]. Therefore, neck dissection should be strongly considered in these cases. Due to high prevalence of extracapsular extension in parotid metastases from cSCC, patients should be aware of possible need to perform total, rather than superficial, parotidectomy and the possibility of sacrifice of the main trunk or branches of the facial nerve. At present, surgical management of regional metastatic disease does not change when treating an immunocompromised patient.
Radiation Therapy
In many cases of locally advanced cSCC, radiation therapy is utilized as part of a curative regimen in either the adjuvant setting, in the case of resectable disease, or definitively, in cases where the tumor is unresectable due to local invasion, location or if the patient is not an operable candidate due to comorbidities. The addition of adjuvant radiation may be considered in the immunocompromised population where it could otherwise be safely omitted. However, even with aggressive combined modality therapy, rates of loco-regional recurrence (LRR) in immunocompromised patients are significantly worse than immunocompetent patients. In a single institution case series of 59 patients, outcomes were dramatically worse in immunocompromised versus immunocompetent cSCC treated with surgery and post-operative radiation therapy (PORT)[72]. In a follow-up study examining a similar question, but with over 200 patients from several institutions, similar results were observed, with dramatically higher rates of LRR following surgery and PORT in immunosuppressed patients (54%) versus immunocompetent patients (17%)(p<0.001) [73]. In both studies tumor characteristics were more unfavorable in immunosuppressed patients, with higher rates of poor differentiation and extracapsular extension (ECE). However, on multivariate analysis, immunosuppression remained a significantly associated with worse LRR. Moreover, the risk of distant metastasis was also significantly higher in immunosuppressed patients (25% vs. 10%).
These retrospective findings would argue for the intensification of adjuvant radiation with cytotoxic chemotherapy, and this is commonly performed off-trial, usually in the context of close or positive margins or ECE, extrapolating from clinical trials of head and neck squamous cell carcinoma [74]. For example, in the aforementioned retrospective study, approximately 14% of patients were treated with concurrent chemoradiation [73]. However, the benefit to adjuvant chemoradiation in cSCC has been called into question by the results of TROG 05.01, a phase III randomized trial, examining loco-regional control in cSCC completely resected followed by PORT or PORT plus weekly carboplatin [75]. In this study, no significant difference was found in freedom from LRR at 2 years between arms (88% vs 89%), nor in Disease Free Survival (DFS) or Overall Survival (OS), although the trial was not powered for the latter two endpoints. However, this study excluded immunosuppressed patients, and used carboplatin instead of cisplatin. The role of adjuvant concurrent chemoradiation specifically in cSCC in SOTRs has not been evaluated.
In patients for whom surgery is not possible or feasible, definitive radiation therapy may be utilized, sometimes with concurrent chemotherapy. The loco-regional recurrence rates following radiation therapy vary between 4% in favorable settings and 30% in the setting of large tumors or other negative prognostic factors (rev. in [76]). In the latter setting, concurrent chemotherapy may be added based on data extrapolated from other disease sites. One small prospective trial of 20 immunocompetent patients and one immunosuppressed patient with locally advanced cSCC on the head and neck treated with weekly platinum and radiation therapy achieved a complete clinical response in slightly over 50% of patients, with an 80% 1 year OS [77]. In the absence of additional prospective data, our bias is to add concurrent chemotherapy to definitive radiation in the locally advanced setting.
Radiation toxicity may be affected by the type of immunosuppressive medication used in SOTRs. Sirolimus, or rapamycin, is part of the post-transplant regimen for many patients. The target of this drug, the mammalian target of rapamycin (mTOR), has been heavily studied as a potential target for radiosensitization in head and neck cancer [78,79]. While a case report demonstrates a dramatic response to a comparatively low dose of radiation in a laryngeal squamous cell carcinoma in a patient on an mTOR inhibitor [80], this patient and two others have suffered toxicities significantly greater than expected [81,82]. In a phase I study of an mTOR inhibitor combined with chemoradiation in head and neck cancer, 5mg day was associated with dose limiting mucositis in 1 of 6 patients, with grade 3 mucositis developing very early on during radiation [83]. Thus, while not definitive data by any means, close monitoring of toxicity is needed when treating with radiotherapy in patients currently on mTOR inhibitors such as everolimus and sirolimus.
Systemic therapy
Systemic therapy remains the primary treatment option for patients with locoregional recurrence without surgical or radiation options and/or metastatic cSCC. Systemic agents evaluated in immunocompetent advanced cSCC include traditional cytotoxic agents (platinum, 5FU, taxane), drugs targeting the Epidermal Growth Factor Receptor (EGFR), and immunotherapy. There is a paucity of data on the efficacy of systemic therapy specifically in SOTRs and to date these systemic therapies have not been studied specifically in this patient population.
Targeted therapy against the epidermal growth factor receptor (EGFR) has been evaluated in advanced cSCC, where EGFR is frequently overexpressed [84]. Cetuximab, an IgG1 monoclonal antibody that targets EGFR, was evaluated in a phase II trial that included 36 patients and it showed a response rate (RR) of 27% and disease control rate (DCR) of 70%, however the duration of activity was short with a median progression free survival (PFS) and OS of 4 and 8 months respectively[84]. Panitumumab, an IgG2 monoclonal antibody against EGFR, was also evaluated prospectively with a RR of 31% in 16 patients[85]. Gefitinib, an oral small molecule inhibitor of EGFR, was tested in advanced disease with zero responses and DCR of 27%[86]. These aforementioned studies were in immunocompetent patients, and only case reports have been published with some response in kidney and heart transplant patients with advanced cSCC. In terms of other potential molecular targets, while certain mutations are shared between HNSCC and cSCC, no direct comparison of the mutational profile in immunocompetent vs. SOTRs cSCCs by whole exome sequencing have been conducted and the question remains as to whether there are any unique mutations in SOTRs that could be targeted.
Immunotherapy has been evaluated in prospective trials in immunocompetent cSCC patients. Interferon alpha in combination with 13-cis-retinoic acid yielded a RR of 68% with 25% achieving a complete response (CR). In combination with cisplatin the overall response rate was 34% with 17% of patients having a CR and those that had a CR had a median duration of response of 34 months. Response rates in patients with locally advanced disease were an impressive 67%[87]. However, due to toxicity, these regimens were never fully integrated into standard practice. More recently, anti-PD-1 monoclonal antibody (mAb) cemiplimab was evaluated in immunocompetent patients with advanced cSCC The RR was an impressive 47% with an estimated one year PFS and OS of 53% and 81% respectively[88]. These results lead to the FDA approval of cemiplimab for advanced cSCC in September 2018.
While efficacious in immunocompetent cSCC patients, risk of organ rejection in SOTRs with checkpoint inhibitors has led to these patients being excluded from trials. Numerous case reports have been published and are summarized in Table 2[89,90,99–108,91,109–112,92–98]. Work done at MD Anderson resulted in a comprehensive review pooling data from 29 patients from published case reports with 10 patients treated locally [113]. In this analysis the majority of patients had melanoma (62%) and had undergone kidney transplant (59%). The analysis included 5 cSCC patients. Forty one percent of patients had graft rejection with 81% of those patients losing their graft despite medical intervention. Graft rejection occurred early after treatment with a median time of 21 days (95% CI 19.3 – 22.8 days) and the majority of tested patients showed acute rejection, T cell-mediated, with 4 out of the 5 patients analyzed for PD-1/PD-L1 showing expression of these co-signaling molecules in the allograft immune microenvironment. While 6 patients received Ipilimumab and Nivolumab (5 Ipilimumab followed by nivolumab and 1 combination therapy) 41% of patients treated with single agent anti-PD-1 and 37% of those just treated with Ipilimumab rejected their graft. This observation differs from initial impressions that the risk was much higher with anti-PD-1/PD-L1 mAb which was based on case reports. Importantly, response rates were comparable to those reported in immunocompetent patients (all 5 cutaneous SCC patients had a response) and did not show a clear correlation with allograft rejection. In regards to immunosuppressive medications, in the MD Anderson series a numerically higher response rate was observed in those that were on single agent prednisone compared to single agent calcineurin or mTOR inhibitors, calcineurin, or combination therapy, 63% vs. 42% respectively, albeit those receiving single agent prednisone had a higher rate of rejection[113]. In our analysis of published case reports, all patients that had their immunosuppression held had allograft rejection.
Table 2.
Age nt | # Cas es | Tumor Type | Tumor ORR | Continued Immunosuppression? | Number of immunosuppressants | Graft Rejection | Median time to Rejection | Mean time to Rejection | Range Time to Rejection |
---|---|---|---|---|---|---|---|---|---|
All | 30 | Melan oma - 15 HCC - 5 SCC - 5 NSCLC - 4 Duode nal adeno −1 |
CR - 3 PR - 9 SD - 5 PD −10 Unkno wn - 4 |
Continued Regimen −7 Decreased Regimen −15 Changed Agents −5 Held −3 |
Three - 2 Two - 12 One - 14 Zero - 2 |
14/30 (47%) | 17.5 days | 28 days | 5 days - 4 mont hs |
PD-1 | 18 | Melan oma - 6 HCC - 6 SCC - 2 NSCLC - 3 Duode nal adeno −1 |
CR - 2 PR - 3 SD - 4 PD −4 Unkno wn - 5 |
Continued Regimen −6 Decreased Regimen −10 Changed Agents −2 Held −0 |
Three - 2 Two - 9 One - 6 Zero - 1 |
9/18 (50%) | 30 days | 33 days | 5 days - 4 mont hs |
CTL A-4 | 7 | Melan oma - 7 | PR - 3 SD - 1 PD −3 |
Continued Regimen −1 Decreased Regimen - 6 |
Two - 1 One - 6 |
2/7 (27%) | 26 days | 26 days | 22 days - 1 mont h |
Bot h | 5 | Melan oma - 3 SCC - 2 |
CR - 1 PR - 2 PD −2 |
Continued Regimen −1 Decreased Regimen - 2 Held - 2 |
Two - 2 One - 1 Zero - 2 |
3/5 (60%) | 8 days | 12 days | 8 – 21 days |
While interesting observations have been made, firm conclusions in SOTRs cannot be made solely based on case reports. Further research is needed for example to better define whether alteration of immunosuppression can modulate response and risk of rejection in these patients. While dialysis can serve as a life-sustaining option for kidney transplant patients, rejection of a lung, heart, or liver is not tenable. Given the significant morbidity and mortality caused by cSCC in SOTRs and impressive efficacy of anti-PD-1/PD-L1 monoclonal antibodies in advanced immunocompetent cSCC, there is a great need for a better understanding of the tumor and graft immune microenvironment and to determine management strategies to maximize efficacy in this patient population while minimizing the risk to the graft. A prospective trial is planned evaluating nivolumab and ipilimumab in kidney transplant patients with malignancy utilizing tacrolimus and prednisone as immunosuppression (NCT03816332). In addition to targeting PD-1:PD-L1, other more tumor specific immune targets may have potential as well as immunotherapies that are injected directly into the tumor. For example, B7-H3 expression has been observed on tumor but not in the immune cell population of the graft in SOTR with cSCC[114].
Conclusion
SOTRs are at an increased risk for cSCC. These tumors are not only more frequent but also at higher risk for LRR and distant metastases in this patient population. Here, we have reviewed the factors contributing to this increase in incidence as well as prevention and management of this disease. The head and neck region is the most common site for this disease and cosmetic and functional considerations make treatment especially challenging. Given the unique nature of these patients, multidisciplinary care is paramount. Further research specifically in this patient population is needed to improve outcomes.
Cutaneous SCCs are more common and more aggressive in transplant recipients
Primary and secondary prevention and patient education can decrease tumor burden
Solid organ transplant patients often require multidisciplinary cancer care
Acknowledgement
K.B. was supported by NIH/NCI T32 CA060397–20.
C.H.L.K. was supported by the Programm zur internen Forschungsforderung Essen (IFORES)
Footnotes
All authors declare no conflicts of interest related to this manuscript submission.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- [1].Karia PS, Han J, Schmults CD. Cutaneous squamous cell carcinoma: Estimated incidence of disease, nodal metastasis, and deaths from disease in the United States, 2012. J Am Acad Dermatol 2013. 10.1016/j.jaad.2012.11.037. [DOI] [PubMed] [Google Scholar]
- [2].Thompson AK, Kelley BF, Prokop LJ, Murad MH, Baum CL. Risk factors for cutaneous squamous cell carcinoma recurrence,metastasis, and disease-specific death: A systematic review and meta-analysis. JAMA Dermatology 2016. 10.1001/jamadermatol.2015.4994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].HW R, MA W, SR F, BM C. Incidence estimate of nonmelanoma skin cancer (keratinocyte carcinomas) in the us population, 2012. JAMA Dermatology 2015. [DOI] [PubMed] [Google Scholar]
- [4].Hartevelt MM, Bouwes Bavinck JN, Kootte AMM, Vermeer BJ, Vandenbroucke JP. Incidence of skin cancer after renal transplantation in the netherlands. Transplantation 1990. 10.1097/00007890-199003000-00006. [DOI] [PubMed] [Google Scholar]
- [5].Jensen P, Hansen S, Moller B, Leivestad T, Pfeifer P, Geiran O, et al. Skin cancer in kidney and heart transplant recipients and different long-term immunosuppressive therapy regimens. J Am Acad Dermatol 1999. 10.1016/S0190-9622(99)70185-4. [DOI] [PubMed] [Google Scholar]
- [6].Lindelöf B, Sigurgeirsson B, Gäbel H, Stern RS. Incidence of skin cancer in 5356 patients following organ transplantation. Br J Dermatol 2000. 10.1046/j.1365-2133.2000.03703.x. [DOI] [PubMed] [Google Scholar]
- [7].Bouwes Bavinck JN, Hardie DR, Green A, Cutmore S, Macnaught A, O’Sullivan B, et al. The risk of skin cancer in renal transplant recipients in Queensland, Australia: A follow-up study. Transplantation 1996. 10.1097/00007890-199603150-00008. [DOI] [PubMed] [Google Scholar]
- [8].Garrett GL, Blanc PD, Boscardin J, Lloyd AA, Ahmed RL, Anthony T, et al. Incidence of and risk factors for skin cancer in organ transplant recipients in the United States. JAMA Dermatology 2017;153 10.1001/jamadermatol.2016.4920. [DOI] [PubMed] [Google Scholar]
- [9].Brantsch KD, Meisner C, Schönfisch B, Trilling B, Wehner-Caroli J, Röcken M, et al. Analysis of risk factors determining prognosis of cutaneous squamous-cell carcinoma: a prospective study. Lancet Oncol 2008. 10.1016/S1470-2045(08)70178-5. [DOI] [PubMed] [Google Scholar]
- [10].Lott DG, Manz R, Koch C, Lorenz RR. Aggressive behavior of nonmelanotic skin cancers in solid organ transplant recipients. Transplantation 2010. 10.1097/TP.0b013e3181ec7228. [DOI] [PubMed] [Google Scholar]
- [11].Zavos G, Karidis NP, Tsourouflis G, Bokos J, Diles K, Sotirchos G, et al. Nonmelanoma skin cancer after renal transplantation: a single-center experience in 1736 transplantations. Int J Dermatol 2011. 10.1111/j.1365-4632.2011.04939.x. [DOI] [PubMed] [Google Scholar]
- [12].Garrett GL, Lowenstein SE, Singer JP, He SY, Arron ST. Trends of skin cancer mortality after transplantation in the United States: 1987 to 2013. J Am Acad Dermatol 2016. 10.1016/j.jaad.2016.02.1155. [DOI] [PubMed] [Google Scholar]
- [13].Chan AW, Fung K, Austin PC, Kim SJ, Singer LG, Baxter NN, et al. Improved keratinocyte carcinoma outcomes with annual dermatology assessment after solid organ transplantation: Population-based cohort study. Am J Transplant 2019. 10.1111/ajt.14966. [DOI] [PubMed] [Google Scholar]
- [14].Hart PH, Norval M. Ultraviolet radiation-induced immunosuppression and its relevance for skin carcinogenesis. Photochem Photobiol Sci 2018. 10.1039/c7pp00312a. [DOI] [PubMed] [Google Scholar]
- [15].Martincorena I, Roshan A, Gerstung M, Ellis P, Van Loo P, Mclaren S, et al. High burden and pervasive positive selection of somatic mutations in normal human skin Europe PMC Funders Group. Science (80- ) 2015;348:880–6. 10.1126/science.aaa6806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Inman GJ, Wang J, Nagano A, Alexandrov LB, Purdie KJ, Taylor RG, et al. The genomic landscape of cutaneous SCC reveals drivers and a novel azathioprine associated mutational signature. Nat Commun 2018. 10.1038/s41467-018-06027-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Ming M, Zhao B, Qiang L, He YY. Effect of immunosuppressants tacrolimus and mycophenolate mofetil on the keratinocyte UVB response. Photochem Photobiol 2015. 10.1111/php.12318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Dziunycz PJ, Lefort K, Wu X, Freiberger SN, Neu J, Djerbi N, et al. The Oncogene ATF3 Is Potentiated by Cyclosporine A and Ultraviolet Light A. J Invest Dermatol 2014;134:1998–2004. 10.1038/jid.2014.77. [DOI] [PubMed] [Google Scholar]
- [19].Zwald FO, Spratt M, Lemos BD, Veledar E, Lawrence C, Marshall Lyon G, et al. Duration of voriconazole exposure: An independent risk factor for skin cancer after lung transplantation. Dermatologic Surg 2012. 10.1111/j.1524-4725.2012.02418.x. [DOI] [PubMed] [Google Scholar]
- [20].Mansh M, Binstock M, Williams K, Hafeez F, Kim J, Glidden D, et al. Voriconazole exposure and risk of cutaneous squamous cell carcinoma, Aspergillus colonization, invasive aspergillosis and death in lung transplant recipients. Am J Transplant 2016. 10.1111/ajt.13431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Gober M, Bashir H, Huang A, Li J, Marshall C, Lee V, et al. Triazole antifungal agents promote UV-DNA damage by increasing oxidative stress. J Invest Dermatol 2015. [Google Scholar]
- [22].Ona K, Oh DH. Voriconazole N-oxide and its ultraviolet B photoproduct sensitize keratinocytes to ultraviolet A. Br J Dermatol 2015. 10.1111/bjd.13862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Pritchett EN, Doyle A, Shaver CM, Miller B, Abdelmalek M, Cusack CA, et al. Nonmelanoma skin cancer in nonwhite organ transplant recipients. JAMA Dermatology 2016. 10.1001/jamadermatol.2016.3328. [DOI] [PubMed] [Google Scholar]
- [24].Strickley JD, Messerschmidt JL, Awad ME, Li T, Hasegawa T, Ha DT, et al. Immunity to commensal papillomaviruses protects against skin cancer. Nature 2019. 10.1038/s41586-019-1719-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Hodges A, Smoller BR. Immunohistochemical comparison of p16 expression in actinic keratoses and squamous cell carcinomas of the skin. Mod Pathol 2002. 10.1097/01.MP.0000032536.48264.D1. [DOI] [PubMed] [Google Scholar]
- [26].Metchnikoff C, Mully T, Singer JP, Golden JA, Arron ST. The 7th edition AJCC staging system for cutaneous squamous cell carcinoma accurately predicts risk of recurrence for heart and lung transplant recipients. J Am Acad Dermatol 2012. 10.1016/j.jaad.2012.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Jambusaria-Pahlajani A, Kanetsky PA, Karia PS, Hwang WT, Gelfand JM, Whalen FM, et al. Evaluation of AJCC tumor staging for cutaneous squamous cell carcinoma and a proposed alternative tumor staging system. JAMA Dermatology 2013. 10.1001/jamadermatol.2013.2456. [DOI] [PubMed] [Google Scholar]
- [28].Karia PS, Jambusaria-Pahlajani A, Harrington DP, Murphy GF, Qureshi AA, Schmults CD. 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. 10.1200/JCO.2012.48.5326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Amin MB, Edge S, Greene F, Byrd DR, Brookland RK, Washington MK, et al. AJCC Cancer Staging Manual Eighth edition. Springer Int Publ 2017. 10.1007/978-3-319-40618-3. [DOI] [Google Scholar]
- [30].Cañueto J, Burguillo J, Moyano-Bueno D, Viñolas-Cuadros A, Conde-Ferreirós A, Corchete-Sánchez LA, et al. Comparing the eighth and the seventh editions of the American Joint Committee on Cancer staging system and the Brigham and Women’s Hospital alternative staging system for cutaneous squamous cell carcinoma: Implications for clinical practice. J Am Acad Dermatol 2019. 10.1016/j.jaad.2018.06.060. [DOI] [PubMed] [Google Scholar]
- [31].Blechman AB, Carucci JA, Stevenson ML. Stratification of Poor Outcomes for Cutaneous Squamous Cell Carcinoma in Immunosuppressed Patients Using the American Joint Committee on Cancer Eighth Edition and Brigham and Womenʼs Hospital Staging Systems. Dermatologic Surg 2019. 10.1097/dss.0000000000001774. [DOI] [PubMed] [Google Scholar]
- [32].OZ F, Skin MB cancer in solid organ transplant recipients: Advances in therapy and management: Part II. Management of skin cancer in solid organ transplant recipients. J Am Acad Dermatol 2011. [Google Scholar]
- [33].Clowers-Webb HE, Christenson LJ, Kim Phillips P, Roenigk RK, Nguyen TH, Weaver AL, et al. Educational Outcomes Regarding Skin Cancer in Organ Transplant Recipients Randomized Intervention of Intensive vs Standard Education. n.d. [DOI] [PubMed] [Google Scholar]
- [34].Petersen B, Wulf HCO, Triguero-Mas M, Philipsen PA, Thieden E, Olsen P, et al. Suppression of TGFβ and Angiogenesis by Type VII Collagen in Cutaneous SCC. J Am Acad Dermatol 2018. 10.1158/1940-6207.CAPR-15-0232. [DOI] [Google Scholar]
- [35].Ulrich C, Jürgensen JS, Degen A, Hackethal M, Ulrich M, Patel MJ, et al. Prevention of non-melanoma skin cancer in organ transplant patients by regular use of a sunscreen: A 24 months, prospective, case-control study. Br J Dermatol 2009. 10.1111/j.1365-2133.2009.09453.x. [DOI] [PubMed] [Google Scholar]
- [36].Christensen SR. Recent advances in field cancerization and management of multiple cutaneous squamous cell carcinomas [version 1; referees: 2 approved]. F1000Research 2018. 10.12688/f1000research.12837.1. [DOI] [PMC free article] [PubMed]
- [37].Perrett CM, McGregor JM, Warwick J, Karran P, Leigh IM, Proby CM, et al. Treatment of post-transplant premalignant skin disease: A randomized intrapatient comparative study of 5-fluorouracil cream and topical photodynamic therapy. Br J Dermatol 2007. 10.1111/j.1365-2133.2006.07616.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Ingham AI, Weightman W. The efficacy and safety of topical 5% 5-fluorouracil in renal transplant recipients for the treatment of actinic keratoses. Australas J Dermatol 2014. 10.1111/ajd.12158. [DOI] [PubMed] [Google Scholar]
- [39].Jirakulaporn T, Endrizzi B, Lindgren B, Mathew J, Lee PK, Dudek AZ. Capecitabine for skin cancer prevention in solid organ transplant recipients. Clin Transplant 2011. 10.1111/j.1399-0012.2010.01348.x. [DOI] [PubMed] [Google Scholar]
- [40].Endrizzi B, Ahmed RL, Ray T, Dudek A, Lee P. Capecitabine to reduce nonmelanoma skin carcinoma burden in solid organ transplant recipients. Dermatologic Surg 2013. 10.1111/dsu.12049. [DOI] [PubMed] [Google Scholar]
- [41].Brown VL, Atkins CL, Ghali L, Cerio R, Harwood CA, Proby CM. Safety and efficacy of 5% imiquimod cream for the treatment of skin dysplasia in high-risk renal transplant recipients: Randomized, double-blind, placebo-controlled trial. Arch Dermatol 2005. 10.1001/archderm.141.8.985. [DOI] [PubMed] [Google Scholar]
- [42].Ulrich C, Bichel J, Euvrard S, Guidi B, Proby CM, Van De Kerkhof PCM, et al. Topical immunomodulation under systemic immunosuppression: Results of a multicentre, randomized, placebo-controlled safety and efficacy study of imiquimod 5% cream for the treatment of actinic keratoses in kidney, heart, and liver transplant patients. Br J Dermatol 2007. 10.1111/j.1365-2133.2007.08269.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Togsverd-Bo K, Halldin C, Sandberg C, Gonzalez H, Wennberg AM, Sørensen SS, et al. Photodynamic therapy is more effective than imiquimod for actinic keratosis in organ transplant recipients: a randomized intraindividual controlled trial. Br J Dermatol 2018. 10.1111/bjd.15884. [DOI] [PubMed] [Google Scholar]
- [44].Joly F, Deret S, Gamboa B, Menigot C, Fogel P, Mounier C, et al. Photodynamic therapy corrects abnormal cancer-associated gene expression observed in actinic keratosis lesions and induces a remodeling effect in photodamaged skin. J Dermatol Sci 2018. 10.1016/j.jdermsci.2018.05.002. [DOI] [PubMed] [Google Scholar]
- [45].Togsverd-Bo K, Omland SH, Wulf HC, Sørensen SS, Hædersdal M. Primary prevention of skin dysplasia in renal transplant recipients with photodynamic therapy: A randomized controlled trial. Am J Transplant 2015. 10.1111/ajt.13358. [DOI] [PubMed] [Google Scholar]
- [46].Willey A, Mehta S, Lee PK. Reduction in the incidence of squamous cell carcinoma in solid organ transplant recipients treated with cyclic photodynamic therapy. Dermatologic Surg 2010. 10.1111/j.1524-4725.2009.01384.x. [DOI] [PubMed] [Google Scholar]
- [47].Dragieva G, Hafner J, Dummer R, Schmid-Grendelmeier P, Roos M, Prinz BM, et al. Topical photodynamic therapy in the treatment of actinic keratoses and Bowen’s disease in transplant recipients. Transplantation 2004. 10.1097/01.TP.0000107284.04969.5C. [DOI] [PubMed] [Google Scholar]
- [48].De Graaf YGL, Kennedy C, Wolterbeek R, Collen AFS, Willemze R, Bavinck JNB. Photodynamic therapy does not prevent cutaneous squamous-cell carcinoma in organtransplant recipients: Results of a randomized-controlled trial. J Invest Dermatol 2006. 10.1038/sj.jid.5700098. [DOI] [PubMed] [Google Scholar]
- [49].Weinstock MA, Bingham SF, Digiovanna JJ, Rizzo AE, Marcolivio K, Hall R, et al. Tretinoin and the prevention of keratinocyte carcinoma (basal and squamous cell carcinoma of the skin): A veterans affairs randomized chemoprevention trial. J Invest Dermatol 2012. 10.1038/jid.2011.483. [DOI] [PubMed] [Google Scholar]
- [50].George R, Weightman W, Russ GR, Bannister KM, Mathew TH. Acitretin for chemoprevention of non-melanoma skin cancers in renal transplant recipients. Australas J Dermatol 2002. 10.1046/j.1440-0960.2002.00613.x. [DOI] [PubMed] [Google Scholar]
- [51].Chen AC, Martin AJ, Choy B, Fernandez-Penas P, Dalziell RA, McKenzie CA, et al. A phase 3 randomized trial of nicotinamide for skin-cancer chemoprevention. N Engl J Med 2015. 10.1056/NEJMoa1506197. [DOI] [PubMed] [Google Scholar]
- [52].Chen AC, Martin AJ, Dalziell RA, McKenzie CA, Lowe PM, Eris JM, et al. A phase II randomized controlled trial of nicotinamide for skin cancer chemoprevention in renal transplant recipients. Br J Dermatol 2016;175:1073–5. 10.1111/bjd.14662. [DOI] [PubMed] [Google Scholar]
- [53].Otley CC, Berg D, Ulrich C, Stasko T, Murphy GM, Salasche SJ, et al. Reduction of immunosuppression for transplant-associated skin cancer: Expert consensus survey. Br J Dermatol 2006. 10.1111/j.1365-2133.2005.07087.x. [DOI] [PubMed] [Google Scholar]
- [54].Jiyad Z, Olsen CM, Burke MT, Isbel NM, Green AC. Azathioprine and Risk of Skin Cancer in Organ Transplant Recipients: Systematic Review and Meta-Analysis. Am J Transplant 2016. 10.1111/ajt.13863. [DOI] [PubMed] [Google Scholar]
- [55].LeBlanc KG, Hughes MP, Sheehan DJ. The role of sirolimus in the prevention of cutaneous squamous cell carcinoma in organ transplant recipients. Dermatologic Surg 2011. 10.1111/j.1524-4725.2011.01973.x. [DOI] [PubMed] [Google Scholar]
- [56].Euvrard S, Morelon E, Rostaing L, Goffin E, Brocard A, Tromme I, et al. Sirolimus and secondary skin-cancer prevention in kidney transplantation. N Engl J Med 2012. 10.1056/NEJMoa1204166. [DOI] [PubMed] [Google Scholar]
- [57].Karia PS, Azzi JR, Heher EC, Hills VM, Schmults CD. Association of sirolimus use with risk for skin cancer in a mixed-organ cohort of solid-organ transplant recipients with a history of cancer. JAMA Dermatology 2016. 10.1001/jamadermatol.2015.5548. [DOI] [PubMed] [Google Scholar]
- [58].Jonas S, Rayes N, Neumann U, Neuhaus R, Bechstein WO, Guckelberger O, et al. De novo malignancies after liver transplantation using tacrolimus-based protocols or cyclosporine-based quadruple immunosuppression with an interleukin-2 receptor antibody or antithymocyte globulin. Cancer 1997. . [DOI] [PubMed] [Google Scholar]
- [59].Moncrieff MD, Shah AK, Igali L, Garioch JJ. False-negative rate of intraoperative frozen section margin analysis for complex head and neck nonmelanoma skin cancer excisions. Clin Exp Dermatol 2015;40:834–8. 10.1111/ced.12743. [DOI] [PubMed] [Google Scholar]
- [60].Chambers KJ, Kraft S, Emerick K. Evaluation of frozen section margins in high-risk cutaneous squamous cell carcinomas of the head and neck. Laryngoscope 2015;125:636–9. 10.1002/lary.24945. [DOI] [PubMed] [Google Scholar]
- [61].Gayre GS, Hybarger CP, Mannor G, Meecham W, Delfanti JB, Mizono GS, et al. Outcomes of excision of 1750 eyelid and periocular skin basal cell and squamous cell carcinomas by modified en face frozen section margin-controlled technique. Int Ophthalmol Clin 2009;49:97–110. 10.1097/IIO.0b013e3181b802ee. [DOI] [PubMed] [Google Scholar]
- [62].Montuno MA, Brown B, Konda S, Motaparthi K. Impact of Mohs Micrographic Surgery on Tumor Staging of Cutaneous Squamous Cell Carcinoma. Dermatologic Surg 2018. 10.1097/dss.0000000000001536. [DOI] [PubMed] [Google Scholar]
- [63].Lansbury L, Bath-Hextall F, Perkins W, Stanton W, Leonardi-Bee J. Interventions for non-metastatic squamous cell carcinoma of the skin: Systematic review and pooled analysis of observational studies. BMJ 2013. 10.1136/bmj.f6153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Rowe DE, Carroll RJ, Day CL. Prognostic factors for local recurrence, metastasis, and survival rates in squamous cell carcinoma of the skin, ear, and lip: Implications for treatment modality selection. J Am Acad Dermatol 1992. 10.1016/0190-9622(92)70144-5. [DOI] [PubMed] [Google Scholar]
- [65].Pugliano-Mauro M, Goldman G. Mohs surgery is effective for high-risk cutaneous squamous cell carcinoma. Dermatologic Surg 2010. 10.1111/j.1524-4725.2010.01576.x. [DOI] [PubMed] [Google Scholar]
- [66].McLaughlin EJ, Miller L, Shin TM, Sobanko JF, Cannady SB, Miller CJ, et al. Rate of regional nodal metastases of cutaneous squamous cell carcinoma in the immunosuppressed patient. Am J Otolaryngol - Head Neck Med Surg 2017. 10.1016/j.amjoto.2017.01.035. [DOI] [PubMed] [Google Scholar]
- [67].Motaparthi K, Kapil JP, Velazquez EF. Cutaneous Squamous Cell Carcinoma: Review of the Eighth Edition of the American Joint Committee on Cancer Staging Guidelines, Prognostic Factors, and Histopathologic Variants. Adv Anat Pathol 2017. 10.1097/PAP.0000000000000157. [DOI] [PubMed] [Google Scholar]
- [68].Tejera-Vaquerizo A, García-Doval I, Llombart B, Cañueto J, Martorell-Calatayud A, Descalzo-Gallego MA, et al. Systematic review of the prevalence of nodal metastases and the prognostic utility of sentinel lymph node biopsy in cutaneous squamous cell carcinoma. J Dermatol 2018. 10.1111/1346-8138.14342. [DOI] [PubMed] [Google Scholar]
- [69].Wu MP, Sethi RK V, Emerick KS. Sentinel lymph node biopsy for high-risk cutaneous squamous cell carcinoma of the head and neck. Laryngoscope 2019. 10.1002/lary.27881. [DOI] [PubMed] [Google Scholar]
- [70].Rotman A, Kerr SJ, Giddings CEB. Elective neck dissection in metastatic cutaneous squamous cell carcinoma to the parotid gland: A systematic review and meta-analysis. Head Neck 2019. 10.1002/hed.25561. [DOI] [PubMed] [Google Scholar]
- [71].Moore BA, Weber RS, Prieto V, El-Naggar A, Holsinger FC, Zhou X, et al. Lymph node metastases from cutaneous squamous cell carcinoma of the head and neck. Laryngoscope 2005. 10.1097/01.mlg.0000173202.56739.9f. [DOI] [PubMed] [Google Scholar]
- [72].Manyam B V, Gastman B, Zhang AY, Reddy CA, Burkey BB, Scharpf J, et al. Inferior outcomes in immunosuppressed patients with high-risk cutaneous squamous cell carcinoma of the head and neck treated with surgery and radiation therapy. J Am Acad Dermatol 2015. 10.1016/j.jaad.2015.04.037. [DOI] [PubMed] [Google Scholar]
- [73].Manyam B V, Garsa AA, Chin R-I, Reddy CA, Gastman B, Thorstad W, et al. A multiinstitutional comparison of outcomes of immunosuppressed and immunocompetent patients treated with surgery and radiation therapy for cutaneous squamous cell carcinoma of the head and neck. Cancer 2017;123:2054–60. 10.1002/cncr.30601. [DOI] [PubMed] [Google Scholar]
- [74].Bernier J, Cooper JS, Pajak TF, Van Glabbeke M, Bourhis J, Forastiere A, et al. Defining risk levels in locally advanced head and neck cancers: A comparative analysis of concurrent postoperative radiation plus chemotherapy trials of the EORTC (#22931) and RTOG (#9501). Head Neck 2005. 10.1002/hed.20279. [DOI] [PubMed] [Google Scholar]
- [75].Porceddu SV, Bressel M, Poulsen MG, Stoneley A, Veness MJ, Kenny LM, et al. Postoperative Concurrent Chemoradiotherapy Versus Postoperative Radiotherapy in High-Risk Cutaneous Squamous Cell Carcinoma of the Head and Neck: The Randomized Phase III TROG 05.01 Trial. J Clin Oncol 2018. 10.1200/JCO.2017.77.0941. [DOI] [PubMed] [Google Scholar]
- [76].Strom T, Harrison LB. Radiotherapy for management of basal and squamous cell carcinoma. Curr Probl Cancer 2015;39:237–47. 10.1016/j.currproblcancer.2015.07.003. [DOI] [PubMed] [Google Scholar]
- [77].Nottage MK, Lin C, Hughes BGM, Kenny L, Smith DD, Houston K, et al. Prospective study of definitive chemoradiation in locally or regionally advanced squamous cell carcinoma of the skin. Head Neck 2017. 10.1002/hed.24662. [DOI] [PubMed] [Google Scholar]
- [78].Ekshyyan O, Rong Y, Rong X, Pattani KM, Abreo F, Caldito G, et al. Comparison of radiosensitizing effects of the mammalian target of rapamycin inhibitor CCI-779 to cisplatin in experimental models of head and neck squamous cell carcinoma. Mol Cancer Ther 2009. 10.1158/1535-7163.MCT-08-1184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Wang Z, Valera JC, Zhao X, Chen Q, Silvio Gutkind J. mTOR co-targeting strategies for head and neck cancer therapy. Cancer Metastasis Rev 2017;36:491–502. 10.1007/s10555-017-9688-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Shinohara ET, Maity A, Jha N, Lustig RA. Sirolimus as a potential radiosensitizer in squamous cell cancer of the head and neck. Head Neck 2009;31:406–11. 10.1002/hed.20898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Manyam B V, Nwizu TI, Rahe ML, Harr BA, Koyfman SA. Early and Severe Radiation Toxicity Associated with Concurrent Sirolimus in an Organ Transplant Recipient with Head and Neck Cutaneous Squamous Cell Carcinoma: A Case Report. Anticancer Res 2015;35:5511–4. [PubMed] [Google Scholar]
- [82].Daste A, de Mones E, Dupin C, François L, Ravaud A, Digue L. m-TOR inhibitor as potential radiosensitizer for head and neck squamous cell carcinoma: A case report of an organ transplant patient and review of the literature. Oral Oncol 2016;62:e1–2. 10.1016/j.oraloncology.2016.08.008. [DOI] [PubMed] [Google Scholar]
- [83].Fury MG, Lee NY, Sherman E, Ho AL, Rao S, Heguy A, et al. A phase 1 study of everolimus + weekly cisplatin + intensity modulated radiation therapy in head-and-neck cancer. Int J Radiat Oncol Biol Phys 2013. 10.1016/j.ijrobp.2013.06.2043. [DOI] [PubMed] [Google Scholar]
- [84].Maubec E, Petrow P, Duvillard P, Laouenan C, Duval X, Lacroix L, et al. Cetuximab as first-line monotherapy in patients with skin unresectable squamous cell carcinoma: Final results of a phase II multicenter study. J Clin Oncol 2010. 10.1200/jco.2010.28.15_suppl.8510. [DOI] [PubMed] [Google Scholar]
- [85].Foote MC, McGrath M, Guminski A, Hughes BGM, Meakin J, Thomson D, et al. Phase II study of single-agent panitumumab in patients with incurable cutaneous squamous cell carcinoma. Ann Oncol 2014. 10.1093/annonc/mdu368. [DOI] [PubMed] [Google Scholar]
- [86].William WN, Feng L, Ferrarotto R, Ginsberg L, Kies M, Lippman S, et al. Gefitinib for patients with incurable cutaneous squamous cell carcinoma: A single-arm phase II clinical trial. J Am Acad Dermatol 2017. 10.1016/j.jaad.2017.07.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Lippman SM, Parkinson DR, Itri LM, Weber RS, Schantz SP, Ota DM, et al. 13-cis-retinoic acid and interferon α −2a: Effective combination. Therapy for advanced squamous cell carcinoma of the skin. J Natl Cancer Inst 1992. 10.1093/jnci/84.4.235. [DOI] [PubMed] [Google Scholar]
- [88].Migden MR, Rischin D, Schmults CD, Guminski A, Hauschild A, Lewis KD, et al. PD-1 blockade with cemiplimab in advanced cutaneous squamous-cell carcinoma. N Engl J Med 2018. 10.1056/NEJMoa1805131. [DOI] [PubMed] [Google Scholar]
- [89].Kittai AS, Oldham H, Cetnar J, Taylor M. Immune Checkpoint Inhibitors in Organ Transplant Patients. J Immunother 2017;40:277–81. 10.1097/CJI.0000000000000180. [DOI] [PubMed] [Google Scholar]
- [90].Kuo JC, Lilly LB, Hogg D. Immune checkpoint inhibitor therapy in a liver transplant recipient with a rare subtype of melanoma: A case report and literature review. Melanoma Res 2018;28:61–4. 10.1097/CMR.0000000000000410. [DOI] [PubMed] [Google Scholar]
- [91].De Toni EN, Gerbes AL. Tapering of Immunosuppression and Sustained Treatment With Nivolumab in a Liver Transplant Recipient. Gastroenterology 2017;152:1631–3. 10.1053/j.gastro.2017.01.063. [DOI] [PubMed] [Google Scholar]
- [92].Rammohan A, Reddy MS, Farouk M, Vargese J, Rela M. Pembrolizumab for metastatic hepatocellular carcinoma following live donor liver transplantation: The silver bullet? Hepatology 2018;67:1166–8. 10.1002/hep.29575. [DOI] [PubMed] [Google Scholar]
- [93].Winkler JK, Gutzmer R, Bender C, Lang N, Zeier M, Enk AH, et al. Safe Administration of An Anti-PD-1 Antibody to Kidney-transplant Patients: 2 Clinical Cases and Review of the Literature. J Immunother 2017;40:341–4. 10.1097/CJI.0000000000000188. [DOI] [PubMed] [Google Scholar]
- [94].Le Fournis S, Gohier P, Urban T, Jeanfaivre T, Hureaux J. Corneal graft rejection in a patient treated with nivolumab for primary lung cancer. Lung Cancer 2016;102:28–9. 10.1016/j.lungcan.2016.10.008. [DOI] [PubMed] [Google Scholar]
- [95].Barnett R, Barta VS, Jhaveri KD. Preserved Renal-Allograft Function and the PD-1 Pathway Inhibitor Nivolumab. N Engl J Med 2017;376:191–2. 10.1056/NEJMc1614298. [DOI] [PubMed] [Google Scholar]
- [96].Jose A, Yiannoullou P, Bhutani S, Denley H, Morton M, Picton M, et al. Renal Allograft Failure After Ipilimumab Therapy for Metastatic Melanoma: A Case Report and Review of the Literature. Transplant Proc 2016;48:3137–41. 10.1016/j.transproceed.2016.07.019. [DOI] [PubMed] [Google Scholar]
- [97].Biondani P, De Martin E, Samuel D. Safety of an anti-PD-1 immune checkpoint inhibitor in a liver transplant recipient. Ann Oncol 2018;29:286–7. 10.1093/annonc/mdx548. [DOI] [PubMed] [Google Scholar]
- [98].Friend BD, Venick RS, McDiarmid S V, Zhou X, Naini B, Wang H, et al. Fatal orthotopic liver transplant organ rejection induced by a checkpoint inhibitor in two patients with refractory, metastatic hepatocellular carcinoma. Pediatr Blood Cancer 2017;64 10.1002/pbc.26682. [DOI] [PubMed] [Google Scholar]
- [99].Schvartsman G, Perez K, Sood G, Katkhuda R, Tawbi H. Immune checkpoint inhibitor therapy in a liver transplant recipient with melanoma. Ann Intern Med 2017;167:361–2. 10.7326/L17-0187. [DOI] [PubMed] [Google Scholar]
- [100].Boils CL, Aljadir DN, Cantafio AW. Use of the PD-1 Pathway Inhibitor Nivolumab in a Renal Transplant Patient With Malignancy. Am J Transplant 2016;16:2496–7. 10.1111/ajt.13786. [DOI] [PubMed] [Google Scholar]
- [101].Gastman BR, Ernstoff MS. Tolerability of immune checkpoint inhibition cancer therapy in a cardiac transplant patient. Ann Oncol Off J Eur Soc Med Oncol 2016;27:2304–5. 10.1093/annonc/mdw293. [DOI] [PubMed] [Google Scholar]
- [102].Ong M, Ibrahim AM, Bourassa-Blanchette S, Canil C, Fairhead T, Knoll G. Antitumor activity of nivolumab on hemodialysis after renal allograft rejection. J Immunother Cancer 2016;4 10.1186/s40425-016-0171-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [103].O TK, K M, Y S, K AO, P RN, AA R, et al. Cardiac allograft rejection as a complication of PD-1 checkpoint blockade for cancer immunotherapy: a case report. Cancer Immunol Immunother 2017;66:45–50. https://doi.org/10.1007/s00262-016-1918-2 LK - https://doi.org/10.1007/s00262-016-1918-2http://sfx.library.uu.nl/utrecht?sid=EMBASE&issn=14320851&id=doi:10.1007%2Fs00262-016-1918-2&atitle=Cardiac+allograft+rejection+as+a+complication+of+PD-1+checkpoint+blockade+for+cancer+immunotherapy%3A+a+case+report&stitle=Cancer+Immunol.+Immunother.&title=Cancer+Immunology%2C+Immunotherapy&volume=66&issue=1&spage=45&epage=50&aulast=Owonikoko&aufirst=Taofeek+K.&auinit=T.K.&aufull=Owonikoko+T.K.&coden=CIIMD&isbn=&pages=45-50&date=2017&auinit1=T&auinitm=K LK - http://sfx.library.uu.nl/utrecht?sid=EMBASE&issn=14320851&id=doi:10.1007%2Fs00262-016-1918-2&atitle=Cardiac+allograft+rejection+as+a+complication+of+PD-1+checkpoint+blockade+for+cancer+immunotherapy%3A+a+case+report&stitle=Cancer+Immunol.+Immunother.&title=Cancer+Immunology%2C+Immunotherapy&volume=66&issue=1&spage=45&epage=50&aulast=Owonikoko&aufirst=Taofeek+K.&auinit=T.K.&aufull=Owonikoko+T.K.&coden=CIIMD&isbn=&pages=45-50&date=2017&auinit1=T&auinitm=K . [DOI] [PMC free article] [PubMed] [Google Scholar]
- [104].Gassmann D, Weiler S, Mertens JC, Reiner CS, Vrugt B, Nägeli M, et al. Liver allograft failure after nivolumab treatment—A Case report with systematic literature research. Transplant Direct 2018;4 10.1097/TXD.0000000000000814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [105].Kwatra V, Karanth N V, Priyadarshana K, Charakidis M. Pembrolizumab for metastatic melanoma in a renal allograft recipient with subsequent graft rejection and treatment response failure: a case report. J Med Case Rep 2017;11:73 10.1186/s13256-017-1229-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [106].Spain L, Higgins R, Gopalakrishnan K, Turajlic S, Gore M, Larkin J. Acute renal allograft rejection after immune checkpoint inhibitor therapy for metastatic melanoma. Ann Oncol Off J Eur Soc Med Oncol 2016;27:1135–7. 10.1093/annonc/mdw130. [DOI] [PubMed] [Google Scholar]
- [107].Morales RE, Shoushtari AN, Walsh MM, Grewal P, Lipson EJ, Carvajal RD. Safety and efficacy of ipilimumab to treat advanced melanoma in the setting of liver transplantation. J Immunother Cancer 2015;3:22 10.1186/s40425-015-0066-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [108].Ranganath HA, Panella TJ. Administration of ipilimumab to a liver transplant recipient with unresectable metastatic melanoma. J Immunother 2015;38:211 10.1097/CJI.0000000000000077. [DOI] [PubMed] [Google Scholar]
- [109].Lipson EJ, Bagnasco SM, Moore J, Jang S, Patel MJ, Zachary AA, et al. Tumor regression and allograft rejection after administration of anti-PD-1. N Engl J Med 2016;374:896–8. 10.1056/NEJMc1509268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].Lipson EJ, Bodell MA, Kraus ES, Sharfman WH. Successful administration of ipilimumab to two kidney transplantation patients with metastatic melanoma. J Clin Oncol 2014;32:e69–71. 10.1200/JCO.2013.49.2314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [111].Alhamad T, Venkatachalam K, Linette GP, Brennan DC. Checkpoint Inhibitors in Kidney Transplant Recipients and the Potential Risk of Rejection. Am J Transplant 2016;16:1332–3. 10.1111/ajt.13711. [DOI] [PubMed] [Google Scholar]
- [112].Herz S, Höfer T, Papapanagiotou M, Leyh JC, Meyenburg S, Schadendorf D, et al. Checkpoint inhibitors in chronic kidney failure and an organ transplant recipient. Eur J Cancer 2016. 10.1016/j.ejca.2016.07.026. [DOI] [PubMed] [Google Scholar]
- [113].Abdel-Wahab N, Safa H, Abudayyeh A, Johnson DH, Trinh VA, Zobniw CM, et al. Checkpoint inhibitor therapy for cancer in solid organ transplantation recipients: An institutional experience and a systematic review of the literature. J Immunother Cancer 2019. 10.1186/s40425-019-0585-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].V V, I OB, B SM, H J, C A, F J, et al. PD-L1, B7-H3, and PD-1 expression in immunocompetent vs. immunosuppressed patients with cutaneous squamous cell carcinoma. Cancer Immunol Immunother 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]