We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Systematic ReviewOpen Accesscc iconby iconnc iconnd icon

Comparative efficacy of cemiplimab versus other systemic treatments for advanced cutaneous squamous cell carcinoma

    Sam Keeping

    *Author for correspondence: Tel.: +1 604 336 3050;

    E-mail Address: sam.keeping@precisionvh.com

    Precision HEOR, Vancouver, BC V6H 3Y4, Canada

    Authors contributed equally

    Search for more papers by this author

    ,
    Yingxin Xu

    Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA

    Authors contributed equally

    Search for more papers by this author

    ,
    Chieh-I Chen

    Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA

    ,
    Shannon Cope

    Precision HEOR, Vancouver, BC V6H 3Y4, Canada

    ,
    Ali Mojebi

    Precision HEOR, Vancouver, BC V6H 3Y4, Canada

    ,
    Andreas Kuznik

    Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA

    , ,
    Dieter Ayers

    Precision HEOR, Vancouver, BC V6H 3Y4, Canada

    ,
    Medha Sasane

    Sanofi, Bridgewater, NJ 08807, USA

    , , ,
    Evan Popoff

    Precision HEOR, Vancouver, BC V6H 3Y4, Canada

    , ,
    Michael L Andria

    Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA

    ,
    Matthew G Fury

    Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA

    , ,
    Eggert Stockfleth

    Department of Dermatology, University of Bochum, 44801 Bochum, Germany

    ,
    Amarnath Challapalli

    Bristol Cancer Institute, University Hospitals Bristol NHS Foundation Trust, Bristol, BS2 8ED, UK

    &
    Chrysalyne D Schmults

    Brigham & Women’s/Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA

    Published Online:https://doi.org/10.2217/fon-2020-0823

    Abstract

    Aim: To estimate the comparative efficacy of cemiplimab, a programmed cell death protein 1 inhibitor, versus EGFR inhibitors, pembrolizumab and platinum-based chemotherapy in terms of overall survival (OS) and progression-free survival. Patients & methods: We performed an indirect treatment comparison of cemiplimab and other available systemic therapies for patients with advanced cutaneous squamous cell carcinoma. Results: Cemiplimab was associated with benefits in OS (hazard ratios range: 0.07–0.52) and progression-free survival (hazard ratios range: 0.30–0.67) versus EGFR inhibitors and pembrolizumab (data from KEYNOTE-629). Cemiplimab was more efficacious versus platinum-based chemotherapy in terms of OS. Conclusion: Cemiplimab may offer improvements in survival for advanced cutaneous squamous cell carcinoma patients compared with existing systemic therapies.

    Lay abstract

    Cutaneous squamous cell carcinoma (CSCC) is a common type of skin cancer that is usually cured by surgery. For some patients, it progresses to a form that cannot be cured by surgery and/or radiotherapy, classified as advanced CSCC. Due to a historical lack of treatment options, other medications have been used in these patients without formal approval. Cemiplimab is a therapy that helps patients fight some types of cancer by boosting the body’s natural defenses. Cemiplimab is the first treatment approved for advanced CSCC in the USA and is also approved in Australia, Brazil, Canada, Europe and Israel. Pembrolizumab, another therapy similar to cemiplimab, has also been investigated and was recently approved in the USA. Currently, there are no clinical trials directly comparing how well cemiplimab works versus existing treatments in patients with advanced CSCC. We performed an indirect treatment comparison using data from clinical studies of cemiplimab and these other systemic therapies. The findings suggest that cemiplimab improves survival relative to other systemic therapies in patients with advanced CSCC.

    Tweetable abstract

    Compared with other systemic treatments, #cemiplimab may increase survival in patients with advanced cutaneous squamous cell carcinoma. #CSCC #cemiplimab #ComparativeEfficacy #advancedCSCC #PDL1 #EGFR #skincancer #cutaneoussquamouscellcarcinoma #nonmelanomaskincancer.

    Cutaneous squamous cell carcinoma (CSCC) is now equivalent in incidence to basal cell carcinoma, each with an estimated incidence of around 1.5 million cases per year in the USA making them by far the most common cancers [1]. Worldwide, reports show a rise in CSCC incidence of 3–7% per year in most countries [2]. The most common cause of CSCC is exposure to ultraviolet rays, which makes those with light skin at highest risk of developing the disease as they lack protective melanin [3,4]. Other risk factors include male gender, advanced age and a compromised immune system [5–9]. Lesions generally form on the parts of the body that are regularly exposed to ultraviolet rays, most frequently the head and neck area, followed by the trunk and the extremities [10,11].

    The current treatment of choice for localized disease is surgical excision, with Mohs micrographic surgery having the highest reported cure rate, although other treatments such as radiotherapy (RT) or curettage/electrodessication may be used in selected cases [12,13]. Prognosis for CSCC patients with localized disease is highly favorable (5-year survival rates are 99% with Mohs micrographic surgery) [12,13]; however, it is estimated that 1% of patients (based on disease-specific mortality rates) go on to develop advanced disease for whom prognosis is often poor [14–16].

    Advanced CSCC is generally defined as either locally advanced disease not amenable to curative surgery/RT (from here on referred to as laCSCC) or metastatic disease (mCSCC), particularly distant metastases or large, multiple and extracapsular nodal disease which has a significant recurrence risk despite lymphadenectomy and radiation [17]. Systemic therapy has generally been used to treat advanced CSCC despite only a small number of prospective uncontrolled studies supporting clinical activity and safety [18,19]. A large retrospective study recently showed that multiple regimens have historically been used in this population with no clear standard and that outcomes are poor with 3-year survival rates being about 30 and 10% for laCSCC and mCSCC patients, respectively [20]. Large clinical trials have not been undertaken among patients with advanced CSCC and a single standard-of-care systemic treatment has not yet been established for this population. This lack of information has hindered the development of definitive treatment guidelines, causing clinicians to rely on evidence of efficacy in other tumor types, such as mucosal squamous cell carcinoma of the head and neck, to guide treatment decisions [19,21]. The limited data that exists for such empiric therapies (e.g., platinum chemotherapy and EGFR inhibitors) have demonstrated limited efficacy in the advanced CSCC population [22–25].

    CSCC tumors harbor a high mutation burden, which in other malignancies has been associated with clinical benefit from antibodies directed against PD-1 immune checkpoint receptor, such as cemiplimab [26–28]. In 2018, cemiplimab (as cemiplimab-rwlc) was granted approval by the US FDA for patients with mCSCC or laCSCC, becoming the first licensed treatment for this specific population [29,30]. The FDA approval for cemiplimab-rwlc was based on data from two clinical trials evaluating patients with advanced CSCC: an open-label, Phase I, multicenter study ( ClinicalTrials.gov: NCT02383212) and a follow-up multicohort, multicenter, Phase II, open-label, single-arm, interventional trial (NCT02760498), where interim results from the two trials showed a centrally reviewed objective response of 47.2% after a median follow-up duration of 8.9 months [29]. Subsequent research based on approximately 1 year of additional follow-up from the Phase II cemiplimab trial indicated that median overall survival (OS) has not been reached and the estimated OS at 24 months was 73.3% (95% CI: 66.1–79.2) [31]. In 2020, pembrolizumab, another PD-1 inhibitor, was approved by the FDA for advanced CSCC based on data from the KEYNOTE-629 study [32].

    No comparative efficacy data currently exists for therapies used to treat advanced CSCC despite being of interest to patients, treating clinicians and reimbursement agencies. The objective of this study was to identify published evidence on the efficacy of systemic treatments used for advanced CSCC to estimate the comparative efficacy of cemiplimab versus other available systemic treatments using three different approaches: an unadjusted (commonly referred to as ‘naive’) comparison, a regression-based simulated treatment comparison (STC) and a matching-adjusted indirect comparison (MAIC) using propensity score weighting. These comparison methods are further described below. Cemiplimab was chosen as the comparator against which other systemic therapies were measured since cemiplimab-rwlc was the only FDA-approved option for CSCC at the time of the analysis and more outcome data is available for patients treated with cemiplimab than for other systemic treatments studied previously. Therefore, physicians and patients may benefit from knowing how cemiplimab performs as compared with other systemic treatment options.

    Patients & methods

    Systematic literature review

    A systematic literature review (SLR) was conducted in May 2019 to identify available studies reporting outcomes of systemic therapy for advanced CSCC. Study eligibility criteria were defined in terms of the population, interventions, comparisons, outcomes and study design structure as outlined in Table 1. Curative surgery was not an intervention of interest since patients in the cemiplimab trials were generally not eligible for this treatment. Furthermore, since there is a lack of consensus over current standard of care for patients with advanced CSCC, no restrictions were placed on the systemic therapies that were eligible for inclusion in the SLR. Relevant studies were identified by searching Excerpta Medica Database (Embase), Medical Literature Analysis and Retrieval System Online (MEDLINE) and Cochrane Central Register of Controlled Trials (CENTRAL) with predefined search strategies (details in the Supplementary data). The database searches were augmented with hand searches of the bibliographies of reviews and meta-analyses along with searches of specific conference proceedings as well as WHO International Clinical Trials Registry Platform (WHO ICTRP) (http://www.who.int/ictrp/search/en/) to identify ongoing and completed clinical trials through May 2019 (Supplementary Table 1). Additional hand searches were conducted in December 2019 to ensure any reports on the identified studies published since the original search were also captured.

    Table 1. Eligibility criteria for the inclusion of studies.
    CriteriaInclusion criteriaExclusion criteria
    PopulationAdult patients with CSCC who:
    • Have locally advanced disease and who are not candidates for surgery, radical RT
    or
    • Have regional metastasis to the lymph nodes or
    • Have distant metastasis.
    Adult patients with other skin cancers (e.g., BCC and melanoma) or noncutaneous SCC (e.g., head and neck) or those with local or locally advanced CSCC who are candidates for treatment with surgery and/or RT
    InterventionsAny systemic interventionSurgical interventions
    ComparatorsAny systemic interventionSurgical interventions
    OutcomesEfficacy outcomes:
    • Overall survival
    • Disease-specific overall survival
    • Progression-free survival
    • Objective response rate
    • Complete response rate
    • Partial response rate
    • Duration of response
    • Duration of disease control
    Health-related quality of life
    Prognostic factors
    Adverse events <grade 3
    Study designInterventional studies (randomized or nonrandomized)
    Observational studies (prospective or retrospective, with ≥10 patients)
    Cross-sectional studies
    Case series with <10 patients
    Case reports
    OthersStudies published in English
    No time limits
    Studies not published in English

    BCC: Basal cell carcinoma; CSCC: Cutaneous squamous cell carcinoma; PICOS: Population, interventions, comparators, outcomes and study design; RT: Radiotherapy; SCC: Squamous cell carcinoma.

    Two reviewers (A Mojebi and S Keeping), working independently, reviewed all abstracts and proceedings identified by the search as well as full-texts of those citations deemed eligible for inclusion according to the population, interventions, comparisons, outcomes and study design criteria. Discrepancies in the decisions between the two reviewers were first reconciled and then a third reviewer (S Cope) was involved to reach a consensus for any remaining discrepancies. The process of study identification and selection was summarized with a Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) flow diagram [33]. Studies involving RT in combination with systemic therapy were excluded since patients in the cemiplimab trials were generally not eligible for curative RT. Studies were also excluded if they did not report the primary study end points of OS or progression-free survival (PFS) via Kaplan–Meier (KM) survival curves as KM data was required to be incorporated in the analysis. Lastly, studies with fewer than 10 patients with advanced CSCC were excluded from this SLR. For the final list of included studies, the initial two reviewers extracted data on study characteristics, interventions, patient characteristics and outcomes. The same process was followed for reconciliation. The Newcastle–Ottawa Scale was used to assess the quality of the studies [34].

    Statistical methods for population-adjusted comparisons between studies

    Since CSCC systemic therapy studies lack untreated control arms, it cannot be determined which component of an observed outcome is attributable to treatment (i.e., the treatment effect) and which part is attributable to prognostic factors or the natural course of disease (i.e., patient study population effect). Therefore, outcome comparisons between studies should be adjusted for differences in their patient populations to enhance accuracy of the comparisons. Population-adjusted comparisons apply statistical methods to individual patient data (IPD) from a given study to estimate how the treatment administered would perform if it had been given to the population from another study. These adjustments are made in order to facilitate a more accurate comparison between the two studies. Thus, in addition to an unadjusted comparison where the observed OS and PFS from each study were compared with those same end points from the cemiplimab trials, two different methods were used for population-adjusted comparisons. These methods (described below) generate estimates of what the OS and PFS would have been if cemiplimab had been included as an arm in the trial/study: regression-based STC and MAIC using propensity score weighting [35].

    A targeted review was conducted to identify which patient and disease factors are prognostic for OS and/or PFS and should therefore be adjusted in the population-adjusted comparisons (see details in the Supplementary Table 2) [7,15,36–60]. Based on the results of the review, two different models were constructed. Prognostic factors included in the core model for the analysis were those reported as statistically significant in at least one study identified in the review: age, disease stage (laCSCC vs mCSCC), tumor grade (well differentiated vs other) and tumor location (head and neck vs other). Additional factors included in the extended model were those not found to be significant or not studied in CSCC but which had been found to be relevant in other tumor types: gender, Eastern Cooperative Oncology Group performance score (0 vs ≥1), prior systemic therapy (yes vs no) and prior RT (yes vs no).

    The reported KM curves for all treatment arms of included studies were first digitized (DigitizeIt; http://www.digitizeit.de/). The algorithm proposed by Guyot et al. was then applied to reconstruct IPD, in other words, survival or censoring times for each patient [61].

    In the STC, the core and extended regression models were first fitted to the IPD from the cemiplimab trials in order to estimate the outcomes of interest (OS and PFS) as a function of the different combinations of prognostic factors. The best fitting models were then used to predict outcomes for cemiplimab in each of the populations from the comparator studies (i.e., OS and PFS if cemiplimab were to be used in the patient population evaluated in the comparator study) and these predicted cemiplimab ‘arms’ were compared with the reported outcomes from each study separately. The goodness of fit was assessed using akaike information criterion (AIC). Bootstrap samples were used to estimate the standard errors of the predicted survival estimates.

    For the MAIC, patients who would not have been eligible for enrolment in the comparator study (based on its inclusion and exclusion criteria) were first removed from the cemiplimab sample. A logistic regression incorporating a propensity score (i.e., the probability of being enrolled in one study vs the other) was then used to estimate weights for the IPD so that, in the MAIC analysis, mean prognostic characteristics of the cemiplimab patients matched those in the populations of comparator studies. These weights were incorporated into the estimation of treatment effects. The distribution of the estimated weights and effective sample sizes are reported in the results section. Note there is no statistic similar to AIC which can be used to compare the fit of different MAIC models, therefore model selection for the MAIC was based on the STC. Relative treatment effects were estimated as hazard ratios (HRs) for OS and PFS by means of Cox regression.

    Results

    Systematic literature review

    Searches were executed on 1 May 2019 and a total of 7682 citations were identified through Embase, MEDLINE and CENTRAL. The PRISMA flow diagram for the study selection process used in the SLR is presented in Figure 1. Searches through other sources resulted in a total of 165 citations categorized as additional records identified through other sources. Overall, 42 citations, corresponding to 27 studies were included [22–25,32,62–97]. The search of the WHO ICTRP found 96 studies, 25 of which were completed (n = 4) or ongoing (n = 21) clinical trials that met the eligibility criteria and were not already captured in the main search. The majority of these trials investigated the use of targeted therapies such as pembrolizumab, nivolumab and panitumumab, as monotherapy or in combination with other therapies. The ongoing trials generally started in or after 2017. None of these 25 studies had any results reported on the WHO ICTRP, ClinicalTrials.gov, or ClinicalTrialsRegister.eu platforms. However, manual searches in December 2019 identified two conference posters reporting on one of these trials (KEYNOTE-629).

    Figure 1. Study flow diagram based on preferred reporting items for systematic reviews and meta-analyses.

    Six conference abstracts were excluded by outcome at this stage.

    Includes citations that also included patients with resectable locally advanced disease (n = 11); citations that also included patients with local CSCC (n = 9).

    §Includes two narrative reviews, one letter, a citation in German and a conference abstract corresponding to an included full-text citation (Nottage et al. [98]) with no additional data.

    CENTRAL: Cochrane Central Register of Controlled Trials; CSCC: Cutaneous squamous carcinoma; Embase: Excerpta Medica Database; MEDLINE: Medical Literature Analysis and Retrieval System Online; RT: Radiotherapy; SLR: Systematic literature review.

    Details of the 27 included studies are presented in Supplementary Table 3. Fourteen of these studies were clinical trials and the remaining 13 were observational studies. None of the clinical trials were randomized or blinded. In terms of stage of disease, the cemiplimab trials recruited both laCSCC and mCSCC patients. Di Monta et al. [67] and Huis In ‘t Veld et al. [74] only included patients with laCSCC, while Gold et al. [22] and Berliner et al. [63] only included patients with mCSCC. Although all subgroups of patients with advanced CSCC were eligible in Foote et al. [69], all 16 patients in this study had regional and/or distant mCSCC. The remaining studies recruited a mix of laCSCC and mCSCC populations. The criteria used to assess whether patients with locally advanced disease were eligible for curative surgery/RT were not always reported and where they were, substantial variation was seen across studies. Overall, the studies in patients with advanced CSCC were of good quality with at least four stars earned on the Newcastle–Ottawa risk of bias assessment scale [34]. Detailed information on the quality assessment of all the included studies can be found in Supplementary Table 4.

    Study selection for comparisons against cemiplimab

    From the 27 studies identified in the SLR, 11 single-arm or retrospective observational studies (representing data from 617 patients) were deemed suitable for inclusion in the comparisons given that they involved systemic drug monotherapy (without RT) and reported OS or PFS via KM survival curves. Analyses were conducted using data from the registrational trial of cemiplimab (i.e., the Phase II study; NCT02760498); therefore, the Phase I trial of cemiplimab (NCT02383212) was not included in the comparisons. Beyond the Phase II cemiplimab trial (n = 193), seven studies evaluated EGFR inhibitors: Gold et al. (n = 39) evaluated erlotinib; Picard et al. [24] (n = 31), Maubec et al. [23] (n = 36) and Peyrade et al. [84] (n = 58) evaluated cetuximab; William et al. [25] (n = 40) evaluated gefitinib; Cavalieri et al. [64] (n = 42) evaluated dacomitinib and Foote et al. (n = 16) evaluated panitumumab. The remaining three studies were Maubec et al. 2019 [96] (n = 39) and Grob et al. [32] (n = 105) who evaluated pembrolizumab and Jarkowski et al. [95] (n = 18) who evaluated chemotherapy with platinum.

    Fifteen studies (Supplementary Table 3) were excluded from the analysis because they fell into either of the following categories: studies that did not provide KM curves of OS or PFS (n = 12), on those including RT in addition to systemic therapy (n = 3). The three studies excluded for inclusion of RT evaluated cisplatin plus RT [80], cetuximab alone or in combination with carboplatin and/or RT [66] and cetuximab plus radiochemotherapy [92].

    A summary of the 11 included studies is presented in Table 2. Although all included studies enrolled patients with advanced CSCC as defined previously (laCSCC or mCSCC), the distribution of laCSCC and mCSCC patients within those populations showed some variation. For example, while almost the entire population consisted of mCSCC patients in Gold et al. [22] (100% mCSCC), Foote et al. [69] (100% mCSCC), William et al. [25] (90% mCSCC) and Maubec et al. [96] (88% mCSCC), other studies had smaller proportions of patients in this category (of note, only patients with distant mCSCC were categorized as ‘metastatic’ in Grob et al. [32]). None of the patients in the Phase II cemiplimab trial were immunosuppressed, which was also the case for Maubec et al. [23], Maubec et al. [96] and Grob et al. [32]; however, almost half of the patients in Picard et al. [24] and over 20% of patients in Cavalieri et al. [64] were immunosuppressed, while other studies did not report on this characteristic. Where reported, tumors were predominantly located in the head and neck area in all studies except Maubec et al. [23] and Jarkowski et al. [95], with 14 and 44% of patients having lesions in those areas, respectively. On the other hand, studies were comparable in terms of patient demographics and performance status: populations were predominantly male across all included studies, with the majority of patients having an Eastern Cooperative Oncology Group performance score of either 0 or 1, where reported. Studies generally had elderly populations with median ages that ranged from 65 years (in Gold et al. [22]) to 86 years (in Picard et al. [24]). Of note, histologic tumor grades were only reported in the IPD of the Phase II cemiplimab trial and in Picard et al. [24], with just over 20% of patients in the former and almost half of the patients in the latter having well-differentiated tumors.

    Table 2. Characteristics of patients and interventions evaluated in the cemiplimab trials and the nine comparator studies included in the analyses.
    Study (trial no./year)TreatmentnAgeMaleECOG PS 0 or 1ImmunosuppressedmCSCCTumor histological gradeTumor locationRef.
            123H & NTrunkExt. 
    Cemiplimab studies
    Phase I trial
    (NCT02383212)
    Cemiplimab, IV (3 mg/kg every 2 weeks; maximum treatment duration: 48 weeks)2672.5
    (52–88)
    21
    (80.8)
    26
    (100)
    0 (0)16
    (61.5)
    2
    (8)
    6
    (23)
    11
    (42)
    19
    (73)
    2
    (7.7)
    5
    (19.2)
     
    Phase II trial
    (NCT02760498)
    Cemiplimab, IV (3 mg/kg every 2 weeks, or 350 mg every 3 weeks)19372
    (38–96)
    161
    (83.4)
    193
    (100)
    0 (0)115
    (59.6)
    41
    (21.2)
    63
    (32.6)
    65
    (33.7)
    128
    (66.3)
    25
    (13)
    40
    (20.7)
     
    Comparator studies
    Gold et al. (2018)Erlotinib, PO (150 mg daily; cycle length: 28 days; UDP)3968
    (45–88)
    34
    (87)
    34
    (87)
    39
    (100)
      31
    (79)
    3
    (8)
    5
    (13)
    [22]
    Jarkowski et al. (2016)Platinum-based chemotherapy2566.4
    (2.8)
    18
    (72)
    6
    (24)
      11
    (44)
    7
    (28)
    3
    (12)
    [95]
    Maubec et al. (2011)Cetuximab, IV (250 mg/m2 weekly; maximum treatment duration: 48 weeks)3679
    (32–95)
    21
    (58)
    28
    (78)
    0 (0)19
    (53)
      5
    (14)
    17
    (47)
    14
    (39)
    [23]
    Picard et al. (2017)Cetuximab, IV (250 mg/m2 weekly; UDP)3186
    (48–96)
    22
    (71)
    20
    (64)
    15
    (48)
    19
    (61)
    15
    (48)
    8
    (26)
    7
    (23)
    22
    (71)
    3
    (10)
    6
    (19)
    [24]
    William et al. (2017)Gefitinib, PO (250 mg daily; cycle length: 28 days; UDP)4067
    (37–95)
    30
    (75)
    36
    (90)
    36
    (90)
      32
    (80)
    2
    (5)
    6
    (15)
    [25]
    Cavalieri et al. (2018), (DACOMINT14)Dacomitinib, PO (Initially 30 mg daily followed by 45 mg daily; cycle length: 28 days; UDP)4277
    (45–91)
    33
    (79)
    41
    (97)
    9 (21)30
    (71)
      28
    (67)
    3
    (7)
    11
    (26)
    [64]
    Foote et al. (2014)Panitumumab, IV (6 mg/kg every 2 weeks; max. treatment duration: 9 cycles)1668
    (47–86)
    14
    (88)
    14
    (88)
    16
    (100)
        [69]
    Maubec et al. (2019), (CARSKIN)Pembrolizumab, IV (200 mg every 3 weeks; max. treatment duration: 24 months)3980
    (43–99)
    31
    (79.5)
    39
    (100)
    0 (0)34
    (88)
      24
    (62)
    3
    (8)
    12
    (31)
    [96]
    Grob et al. (2019), (KEYNOTE-629)Pembrolizumab, IV (200 mg every 3 weeks; max. treatment duration: 24 months, or UDP)10572
    (29–95)
    80
    (76.2)
    105
    (100)
    0 (0)≥58
    (55.2)§
        [32]
    Peyrade et al. (2018)Cetuximab (dose and schedule were not reported)5883.2
    (47–96)
    38
    (66)
    32
    (55)
    20
    (34.5)
        [84]

    Values for age are reported as median (range). All other values are reported as n (%).

    Per eligibility criteria.

    Mean (standard deviation) was reported.

    §Only patients with distant metastasis were included in the ‘metastatic’ category in Grob et al. [32] (KEYNOTE-629). Therefore, there may have been some patients with regional mCSCC who were not included in that category.

    Indicate that the characteristic was not reported in the study.

    CSCC: Cutaneous squamous cell carcinoma; Ext: Extremities; IV: Intravenous; ECOG PS: Eastern Cooperative Oncology Group performance score; H & N: Head and neck; mCSCC: Metastatic CSCC; PO: Per os (oral); UDP: Until disease progression.

    There were also some differences in terms of the prior treatment experience of enrolled patients. Similar to the Phase II cemiplimab trial, Grob et al. [32] excluded patients if they had received prior treatment with PD-1 or programmed death-ligand 1 inhibitors, with no restriction for prior chemotherapy or EGFR inhibitors. Patients in Cavalieri et al. [64] had not received prior EGFR inhibitors. Gold et al. [22] and William et al. [25] had the same restriction and also could have only received up to one prior chemotherapy regimen (i.e., patients were receiving their first- or second-line of systemic treatment). The population in Peyrade et al. [84] had not received prior EGFR inhibitors or chemotherapy in the advanced stage of the disease. Maubec et al. [23] and Maubec et al. (2019) [96] had the most restrictive eligibility criteria and only included patients who were receiving their first-line of systemic treatment. In Picard et al. [24], only three patients (10%) had received prior chemotherapy, which was in conjunction with surgery and RT; the remaining patients had never received chemotherapy prior to study entry. Furthermore, patients who had prior treatment experience with EGFR inhibitors were excluded according to the eligibility criteria of this study. It was, therefore, concluded that the population in Picard et al. [24] were receiving their first-line of systemic treatment in the advanced stage. Jarkowski et al. [96] and Foote et al. [69] did not have any restrictions for prior treatment experience.

    Reported outcomes

    Both OS and PFS were reported across all included studies with the exception of Picard et al. [24], which did not provide a KM curve for PFS. Figures 2 & 3 present the reported KM curves from included studies for OS and PFS, respectively. Median OS was not reached for cemiplimab in the Phase II clinical trial (after median follow-up duration of 11.9 months). In Jarkowski et al. [95], chemotherapy with platinum was associated with a median OS of 15.1 months. For treatment with cetuximab, median OS was reported as 13 months (range 1–36) and 17.5 months (95% CI: 9.4–43.1) in Picard et al. [24] and Peyrade et al. [84], respectively, and a mean OS of 8.1 months (95% CI: 6.9–9.3) was reported in Maubec et al. [23]. Survival with gefitinib and erlotinib were comparable, with a median OS of 12.9 months (95% CI: 8.5–25.0) in William et al. [25] for the former and 14.9 months (95% CI: 11.7–25.2) in Gold et al. [22] for the latter. Median OS was 11 months with both dacomitinib (Cavalier et al. [64]; 95% CI: 9–13) and panitumumab (Foote et al. [69]). Median OS was not reached with pembrolizumab in either Maubec et al. 2019 [96] or Grob et al. [32].

    Figure 2. Kaplan–Meier curves for overall survival extracted from included studies.

    OS: Overall survival.

    Figure 3. Kaplan–Meier curves for progression-free survival extracted from included studies.

    PFS: Progression-free survival.

    An estimated median PFS of 18.4 months (95% CI: 9.2–not evaluable) was reported for the Phase II cemiplimab trial. Median PFS was 9.8 months for platinum-based chemotherapy in Jarkowski et al. [95]. Treatment with cetuximab resulted in a median PFS of 4.1 months (95% CI: 1.7–5.0) in Maubec et al. (2011) [23], 9 months (range 0–36) in Picard et al. [24] and 9.7 months (95% CI: 4.8–43.4) in Peyrade et al. [84]. PFS estimates for gefitinib and erlotinib were comparable, with medians of 3.8 months (95% CI: 2.2–5.7) in William et al. [25] for the former and 4.8 months (95% CI: 3.6–7.1) in Gold et al. [22] for the latter. Similar to OS, median PFS was comparable for dacomitinib (Cavalieri et al. [64]) and panitumumab (Foote et al. [69]) and was reported as 6 months (95% CI: 4–9) and 8 months, respectively. Median PFS with pembrolizumab was 14.2 and 6.9 months in Maubec et al. [96] and Grob et al. [32], respectively.

    Indirect comparisons

    For OS, the best fitting model in terms of AIC was the extended model for all comparisons except chemotherapy with platinum, where the core model provided a better fit. For PFS, the core model was the best fit for all comparisons.

    Figure 4 shows the naive (unadjusted), STC and MAIC comparisons of cemiplimab versus the EGFR inhibitors. Cemiplimab was superior to EGFR inhibitors in all comparisons and under all methods (except only three HR estimates that were not statistically significant, but still favored cemiplimab), with estimated HRs ranging from 0.30 to 0.67 for PFS and from 0.07 to 0.47 for OS, indicating cemiplimab is protective against disease progression and death, respectively, as compared with EGFR inhibitors.

    Figure 4. Forest plot of hazard ratios of overall survival and progression-free survival survival for cemiplimab versus epidermal growth factor receptor inhibitors.

    MAIC: Matching-adjusted indirect comparison; OS: Overall survival; PFS: Progression-free survival; STC: Simulated treatment comparison.

    Figure 5 shows the results of the comparisons with the remaining interventions (chemotherapy with platinum and pembrolizumab). Cemiplimab was again more efficacious than both treatments in terms of OS (HRs ranging from 0.17 to 0.52). It was also more efficacious in terms of PFS when compared with pembrolizumab (HRs ranging from 0.49 to 0.55), using data from Grob et al. [32]; however, 95% CIs included one when using data from Maubec et al. [96]. Estimated 95% CIs also included one for the PFS comparison versus chemotherapy with platinum.

    Figure 5. Forest plot of hazard ratios of overall survival and progression-free survival survival for cemiplimab versus other interventions.

    MAIC: Matching-adjusted indirect comparison; OS: Overall survival; PFS: Progression-free survival; STC: Simulated treatment comparison.

    Discussion

    This study represents the first attempt to estimate the comparative efficacy of available systemic monotherapies for CSCC. Published OS and PFS data were compared between cemiplimab and other treatments using three different approaches; an unadjusted/naïve comparison, an STC and an MAIC. The latter two approaches mathematically adjust data for differences in study populations. These available adjusted and unadjusted data indicate cemiplimab is protective against disease progression and death as compared with EGFR inhibitors (HRs ranging from 0.30 to 0.67 and from 0.07 to 0.47 for PFS and OS, respectively). Cemiplimab also showed better OS as compared with platinum chemotherapy (HRs ranging from 0.17 to 0.30) and pembrolizumab (HRs ranging from 0.21 to 0.52) and better PFS when data from the largest (n = 105) study of pembrolizumab was used (HRs ranging from 0.49 to 0.55).

    Although CSCC has been estimated to cause deaths comparable in number to melanoma in some regions of the USA [99], it is usually easily cured with surgical removal and locally advanced and metastatic forms of the disease comprise only a small subset of all CSCC. Cohort studies indicate that approximately 80% of CSCC patients who die from disease do not have internal metastases and succumb to local and/or nodal disease [14,15]. This has historically led to relatively few referrals for systemic therapy, an under-recognition of advanced CSCC as a significant disease entity and a relatively slow evolution of therapeutic standards for advanced disease failing surgery and RT. However, since CSCC is known to be a highly immunologically mediated cancer (given the elevated incidence of aggressive CSCC in patients with organ transplants or chronic lymphocytic leukemia), immunotherapy was considered worthy of investigation in advanced CSCC with trials leading to the approval of cemiplimab and pembrolizumab. Population-adjusted treatment comparisons of cemiplimab versus other systemic monotherapies were used in this analysis since no controlled comparative trial exists for advanced CSCC. The data herein supports cemiplimab therapy over other available systemic options for laCSCC and mCSCC not amenable to cure with surgery and/or RT.

    The primary limitation of this study is the low number of comparator studies available and the small numbers of patients enrolled in some studies. The latter was despite the fact that studies with fewer than ten patients were excluded from the current review; of note, many such studies were included in a recent SLR that identified a number of case studies evaluating PD-1 inhibitors in patients with nonmelanoma skin cancers [28]. PFS was not found to be significantly different between cemiplimab and platinum chemotherapy (Jarkowski et al. [95], n = 18) or when using data from the smaller study of pembrolizumab (Maubec et al. [2019] [96], n = 39). Given the small number of patients, these data may have been underpowered to detect a difference between therapies. This is supported by the statistically superior PFS seen with cemiplimab when using data from the larger study of pembrolizumab (Grob et al. [32], n = 105). Since trials directly comparing systemic treatment options do not exist for CSCC, treatment comparisons adjusted for differences in study populations had to be undertaken. The methods used for both the STC and MAIC follow best practice for population-adjusted comparisons and can be considered the best attempt to account for between-study differences given the challenge of evaluating comparative efficacy on the basis of single-arm clinical trials [35]. However, as with any analysis of single-arm trials, it is uncertain whether any unknown or unmeasured prognostic factors missing from the models may have influenced the outcomes of interest. For example, many trials did not report information regarding tumor differentiation despite the established importance of this prognostic factor in both earlier stage and advanced CSCC. Another example is that responses were centrally reviewed or assessed by independent radiologists in some of the studies (e.g., the Phase II cemiplimab trial, Maubec et al. [2011] [23], Foote et al. [69]) while this was not the case for others; while not expected to affect OS, this difference in response assessment may impact the time when disease progression is actually diagnosed (i.e., a PFS event) and, therefore, can influence the estimated PFS HRs. Additionally, analyses could not account for population differences between the Phase II cemiplimab trial and the comparator studies in cases where certain patients were excluded from the former but were present in the latter; for example, nearly half of the population in Picard et al. [24] were immunosuppressed, whereas such patients were not eligible in the cemiplimab trials. Another limitation is that efficacy analyses were not based on the intention-to-treat population in Cavalieri et al. [64], as there were two patients in this study who withdrew consent after receiving less than 1 month of dacomitinib and were excluded from the analyses. Efficacy estimates would have been lower for this study if these two patients had been included; however, hazards were unlikely to be greatly altered given the 42 remaining patients analyzed. A final limitation is that in the Maubec et al. [96] study (ASCO 2019 conference poster), the numbers of patients at risk were identical at all time points for both OS and PFS, suggesting that patients may have been erroneously censored for OS at the time of progression, thus underestimating OS. The OS comparisons of cemiplimab with pembrolizumab using data from Maubec et al. [96] should therefore be interpreted with caution.

    Conclusion

    Cemiplimab demonstrates improvements in OS that are greater than that observed in studies of other systemic treatments. Results regarding PFS were equivocal. Improvement in PFS with cemiplimab was observed in comparisons with EGFR inhibitors and one of the pembrolizumab trials (KEYNOTE-629). However, PFS with cemiplimab was not greater than that observed with platinum-based chemotherapy. Taken together, these analyses indicate that cemiplimab is the systemic therapy with the strongest evidence of clinical benefit to support its use in patients with advanced CSCC.

    Future perspective

    Until recently, there was no approved systemic therapy for patients with advanced CSCC. Previous studies have demonstrated that cemiplimab provides substantial antitumor activity, durable response and acceptable safety profile in patients with CSCC. As cemiplimab was the first treatment approved for advanced CSCC in the USA and is also approved in Australia, Brazil, Canada, Europe and Israel, we anticipate that cemiplimab will play a central role in the treatment of those patients over the next 5–10 years. Ongoing studies in patients with CSCC will provide additional data on pre-operative cemiplimab; cemiplimab alone and in combination with RP1, an investigational genetically modified herpes simplex type 1 virus; cemiplimab in allogeneic hematopoietic stem cell transplantation/solid organ transplantation recipients; cemiplimab before and after surgery; cemiplimab in combination with other agents such as AST-008, a TLR9 agonist and ASP-1929, an anti-EGFR; and real-world use and benefit of cemiplimab, particularly in comparison with other emerging immunotherapies such as pembrolizumab. As clinical evidence supporting cemiplimab matures and real-world evidence is collected, we anticipate that cemiplimab will continue to be a mainstay treatment for patients with CSCC requiring systemic therapy.

    Summary points
    • Cemiplimab is a programmed cell death protein 1 inhibitor approved in the USA (as cemiplimab-rwlc), Australia, Brazil, Canada, Europe and Israel for the treatment of advanced cutaneous squamous cell carcinoma (CSCC).

    • We aimed to systematically identify evidence on efficacy of available systemic treatments for patients with advanced CSCC and to estimate the comparative efficacy of cemiplimab versus other systemic inhibitors in terms of overall survival (OS) and progression-free survival (PFS).

    • A systematic literature review of Embase, MEDLINE and Cochrane Central Register of Controlled Trials conducted in May 2019 identified studies reporting OS or PFS of systemic therapy for patients with locally advanced and/or metastatic CSCC.

    • The effect of treatment on OS or PFS for cemiplimab versus other systemic monotherapies were reported via an unadjusted comparison, a regression-based simulated treatment comparison and a matching-adjusted indirect comparison using propensity score weighting.

    • A total of 27 systemic therapy studies were identified. Of these, 11 studies of systemic monotherapy reporting OS or PFS were included in the comparisons, representing data from 617 patients. Cemiplimab data was based a Phase II trial ( ClinicalTrials.gov: NCT02760498; n = 193). Comparator treatments included EGFR inhibitors, pembrolizumab and platinum-based chemotherapy.

    • Cemiplimab was associated with improved OS (hazard ratios ranging from 0.07 to 0.47) and PFS (hazard ratios ranging from 0.30 to 0.67) versus EGFR inhibitors and pembrolizumab (data from the largest trial, KEYNOTE-629). Cemiplimab was also more efficacious in terms of OS relative to platinum-based chemotherapy (hazard ratios ranging from 0.17 to 0.30).

    • These analyses suggest that cemiplimab may offer improvements in survival for advanced CSCC compared with existing systemic therapies.

    Supplementary data

    To view the supplementary data that accompany this paper please visit the journal website at: www.futuremedicine.com/doi/suppl/10.2217/fon-2020-0823

    Author contributions

    Conception/design was performed by S Keeping, S Cope, A Mojebi, D Ayers, E Popoff, C Chen, A Kuznik, Y Xu, M Sasane, G Konidaris, R Allen and T-M-T Huynh. Provision of study material or patients was provided by C Chen, A Kuznik, Y Xu, M Sasane, G Konidaris, R Allen, T-M-T Huynh, M Freeman, ML Andria, MG Fury, K Singh, E Stockfleth, A Challapalli and CD Schmults. Collection and/or assembly of data was performed by S Keeping, S Cope, A Mojebi, D Ayers, E Popoff and CD Schmults. Data analysis and interpretation were performed by S Keeping, S Cope, A Mojebi, D Ayers, E Popoff, C Chen, A Kuznik, Y Xu, M Sasane, G Konidaris, R Allen, T-M-T Huynh, M Freeman, ML Andria, MG Fury, K Singh, E Stockfleth, A Challapalli and CD Schmults. Manuscript writing was performed by S Keeping. Final approval of manuscript was provided by S Keeping, S Cope, A Mojebi, D Ayers, E Popoff, CI Chen, A Kuznik, Y Xu, M Sasane, G Konidaris, R Allen, T-M-T Huynh, M Freeman, ML Andria, MG Fury, K Singh, E Stockfleth, A Challapalli and CD Schmults.

    Acknowledgments

    The following people reviewed and provided editorial comments on the manuscript: V Mastey, T Michelini, J Lee and A Seluzhtsky.

    Financial & competing interests disclosure

    S Keeping, S Cope, A Mojebi and D Ayers are employees of Precision Xtract who received funding to produce this work. E Popoff was an employee of Precision Xtract at the time of conducting the analyses. C Chen, A Kuznik and Y Xu are employees and stockholders of Regeneron Pharmaceuticals, Inc. MG Fury is an employee and stockholder of Regeneron Pharmaceuticals, Inc.; he also reports involvement in patents and/or intellectual property for Regeneron Pharmaceuticals, Inc. M Sasane and K Singh are employees and stockholders of Sanofi. G Konidaris, R Allen and T-M-T Huynh are employees of Sanofi. M Freeman reports no conflict of interest. A Challapalli reports an advisory role with Regeneron Pharmaceuticals, Inc. ML Andria is a former employee and stockholder of Regeneron Pharmaceuticals, Inc. E Stockfleth reports an advisory role with Regeneron Pharmaceuticals, Inc. CD Schmults reports advisory roles for Regeneron Pharmaceuticals, Inc. and Castle Biosciences. She has received clinical trial research funding and an honorarium from Regeneron Pharmaceuticals, Inc. and clinical trial research funding from Genentech, Novartis, Merck and Castle Biosciences. She has received an unrestricted research grant from Genentech. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    Administrative support was provided by C Hoover of Prime Global (Knutsford, UK); this was funded by Regeneron Pharmaceuticals, Inc. and Sanofi Genzyme according to Good Publication Practice guidelines (https://www.ismpp.org/gpp3 ).

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Papers of special note have been highlighted as: • of interest

    References

    • 1. Rogers HW, Weinstock MA, Feldman SR, Coldiron BM. Incidence estimate of nonmelanoma skin cancer (keratinocyte carcinomas) in the U.S. population, 2012. JAMA Dermatol. 151(10), 1081–1086 (2015).
    • 2. Lucas R, McMichael T, Smith W, Armstrong B. Solar Ultraviolet Radiation: Global Burden of Disease from Solar Ultraviolet Radiation. Environmental Burden of Disease Series, No. 13. WHO, Geneva, Switzerland (2006).
    • 3. Fears TR, Scotto J. Estimating increases in skin cancer morbidity due to increases in ultraviolet radiation exposure. Cancer Invest. 1(2), 119–126 (1983).
    • 4. Johnson TM, Rowe DE, Nelson BR, Swanson NA. Squamous cell carcinoma of the skin (excluding lip and oral mucosa). J. Am. Acad. Dermatol. 26(3 Pt 2), 467–484 (1992).
    • 5. Ducroux E, Martin C, Bouwes Bavinck JN et al. Risk of aggressive skin cancers after kidney retransplantation in patients with previous posttransplant cutaneous squamous cell carcinomas: a retrospective study of 53 cases. Transplantation 101(4), e133–e141 (2017).
    • 6. Lindelof B, Jarnvik J, Ternesten-Bratel A, Granath F, Hedblad MA. Mortality and clinicopathological features of cutaneous squamous cell carcinoma in organ transplant recipients: a study of the Swedish cohort. Acta Derm. Venereol. 86(3), 219–222 (2006).
    • 7. Manyam BV, Garsa AA, Chin RI et al. A multi-institutional comparison of outcomes of immunosuppressed and immunocompetent patients treated with surgery and radiation therapy for cutaneous squamous cell carcinoma of the head and neck. Cancer 123(11), 2054–2060 (2017).
    • 8. McLaughlin EJ, Miller L, Shin TM et al. Rate of regional nodal metastases of cutaneous squamous cell carcinoma in the immunosuppressed patient. Am. J. Otolaryngol. 38(3), 325–328 (2017).
    • 9. Velez NF, Karia PS, Vartanov AR, Davids MS, Brown JR, Schmults CD. Association of advanced leukemic stage and skin cancer tumor stage with poor skin cancer outcomes in patients with chronic lymphocytic leukemia. JAMA Dermatol. 150(3), 280–287 (2014).
    • 10. Gray DT, Suman VJ, Su WP, Clay RP, Harmsen WS, Roenigk RK. Trends in the population-based incidence of squamous cell carcinoma of the skin first diagnosed between 1984 and 1992. Arch. Dermatol. 133(6), 735–740 (1997).
    • 11. Mourouzis C, Boynton A, Grant J et al. Cutaneous head and neck SCCs and risk of nodal metastasis – UK experience. J. Craniomaxillofac. Surg. 37(8), 443–447 (2009).
    • 12. 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 347, f6153 (2013).
    • 13. Hollestein LM, De Vries E, Nijsten T. Trends of cutaneous squamous cell carcinoma in the Netherlands: increased incidence rates, but stable relative survival and mortality 1989–2008. Eur. J. Cancer 48(13), 2046–2053 (2012).
    • 14. Brantsch KD, Meisner C, Schonfisch B et al. Analysis of risk factors determining prognosis of cutaneous squamous-cell carcinoma: a prospective study. Lancet Oncol. 9(8), 713–720 (2008).
    • 15. Schmults CD, Karia PS, Carter JB, Han J, Qureshi AA. Factors predictive of recurrence and death from cutaneous squamous cell carcinoma: a 10-year, single-institution cohort study. JAMA Dermatol. 149(5), 541–547 (2013).
    • 16. Clayman GL, Lee JJ, Holsinger FC et al. Mortality risk from squamous cell skin cancer. J. Clin. Oncol. 23(4), 759–765 (2005).
    • 17. Veness MJ, Morgan GJ, Palme CE, Gebski V. Surgery and adjuvant radiotherapy in patients with cutaneous head and neck squamous cell carcinoma metastatic to lymph nodes: combined treatment should be considered best practice. Laryngoscope 115(5), 870–875 (2005).
    • 18. Cranmer LD, Engelhardt C, Morgan SS. Treatment of unresectable and metastatic cutaneous squamous cell carcinoma. Oncologist 15(12), 1320–1328 (2010).
    • 19. Stratigos A, Garbe C, Lebbe C et al. Diagnosis and treatment of invasive squamous cell carcinoma of the skin: European consensus-based interdisciplinary guideline. Eur. J. Cancer 51(14), 1989–2007 (2015).
    • 20. Cowey CL, Robert NJ, Espirito JL et al. Clinical outcomes among unresectable, locally advanced and metastatic cutaneous squamous cell carcinoma patients treated with systemic therapy. Cancer Med. 9(20), 7381–7387 (2020). • Provides historic benchmarks for outcomes in patients with advanced cutaneous squamous cell carcinoma (CSCC) prior to FDA approval of cemiplimab.
    • 21. Palyca P, Koshenkov VP, Mehnert JM. Developments in the treatment of locally advanced and metastatic squamous cell carcinoma of the skin: a rising unmet need. Am. Soc. Clin. Oncol. Educ. Book 89(15), 7536–7549 (2014).
    • 22. Gold KA, Kies MS, William WN Jr, Johnson FM, Lee JJ, Glisson BS. Erlotinib in the treatment of recurrent or metastatic cutaneous squamous cell carcinoma: a single-arm Phase II clinical trial. Cancer 124(10), 2169–2173 (2018).
    • 23. Maubec E, Petrow P, Scheer-Senyarich I et al. Phase II study of cetuximab as first-line single-drug therapy in patients with unresectable squamous cell carcinoma of the skin. J. Clin. Oncol. 29(25), 3419–3426 (2011).
    • 24. Picard A, Pedeutour F, Peyrade F et al. Association of oncogenic mutations in patients with advanced cutaneous squamous cell carcinomas treated with cetuximab. JAMA Dermatol. 153(4), 291–298 (2017).
    • 25. William WN Jr, Feng L, Ferrarotto R et al. Gefitinib for patients with incurable cutaneous squamous cell carcinoma: a single-arm Phase II clinical trial. J. Am. Acad. Dermatol. 77(6), 1110–1113 e1112 (2017).
    • 26. Alexandrov LB, Nik-Zainal S, Wedge DC et al. Signatures of mutational processes in human cancer. Nature 500(7463), 415–421 (2013).
    • 27. Pickering CR, Zhou JH, Lee JJ et al. Mutational landscape of aggressive cutaneous squamous cell carcinoma. Clin. Cancer Res. 20(24), 6582–6592 (2014).
    • 28. Choi FD, Kraus CN, Elsensohn AN et al. Programmed cell death 1 protein and programmed death-ligand 1 inhibitors in the treatment of nonmelanoma skin cancer: a systematic review. J. Am. Acad. Dermatol. 82(2), 440–459 (2020). • Provides evidence for programmed cell death protein 1 inhibitor and programmed death-ligand 1 inhibitors in the treatment of nonmelanoma skin cancers.
    • 29. Regeneron Pharmaceuticals, Inc. FDA label for cemiplimab (LIBTAYO®). (2020). https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/761097s000lbl.pdf
    • 30. US FDA. FDA approves first treatment for advanced form of the second most common skin cancer. (2020). https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm622044.htm
    • 31. Rischin D, Khushalani NI, Schmults CD et al. Phase II study of cemiplimab in patients (pts) with advanced cutaneous squamous cell carcinoma (CSCC): longer follow-up. J. Clin. Oncol. 38(Suppl. 15), 10018–10018 (2020).
    • 32. Grob J, Gonzalez Mendoza R, Basset-Seguin N et al. LBA72 pembrolizumab for recurrent/metastatic cutaneous squamous cell carcinoma (cSCC): efficacy and safety results from the Phase II KEYNOTE-629 study. Ann. Oncol. 30(Suppl. 5), v908 (2019).
    • 33. Moher D, Liberati A, Tetzlaff J, Altman DG, Group P. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. J. Clin. Epidemiol. 62(10), 1006–1012 (2009).
    • 34. Wells GA, Shea B, O'Connell D, Ottawa Hospital Research Institute et al. The Newcastle–Ottawa Scale (NOS) for assessing the quality of nonrandomised studies in meta-analyses. (2020). http://www.ohri.ca/programs/clinical_epidemiology/oxford.asp
    • 35. National Institute for Health and Care Excellence. NICE DSU Technical support document 18: methods for population-adjusted indirect comparisons in submissions to NICE. (2018). http://nicedsu.org.uk/wp-content/uploads/2018/08/Population-adjustment-TSD-FINAL-ref-rerun.pdf
    • 36. Bachar G, Mizrachi A, Rabinovics N et al. Prognostic factors in metastatic cutaneous squamous cell carcinoma of the head and neck. Ear Nose Throat J. 95(10–11), E32–E36 (2016).
    • 37. Brinkman JN, Hajder E, van der Holt B, Den Bakker MA, Hovius SE, Mureau MA. The effect of differentiation grade of cutaneous squamous cell carcinoma on excision margins, local recurrence, metastasis and patient survival: a retrospective follow-up study. Ann. Plast. Surg. 75(3), 323–326 (2015).
    • 38. Carter JB, Johnson MM, Chua TL, Karia PS, Schmults CD. Outcomes of primary cutaneous squamous cell carcinoma with perineural invasion: an 11-year cohort study. JAMA Dermatol. 149(1), 35–41 (2013).
    • 39. Cheng J, Yan S. Prognostic variables in high-risk cutaneous squamous cell carcinoma: a review. J. Cutan. Pathol. 43(11), 994–1004 (2016).
    • 40. Ch'ng S, Maitra A, Allison RS et al. Parotid and cervical nodal status predict prognosis for patients with head and neck metastatic cutaneous squamous cell carcinoma. J. Surg. Oncol. 98(2), 101–105 (2008).
    • 41. Ch'ng S, Maitra A, Lea R, Brasch H, Tan ST. Parotid metastasis--an independent prognostic factor for head and neck cutaneous squamous cell carcinoma. J. Plast. Reconstr. Aesthet. Surg. 59(12), 1288–1293 (2006).
    • 42. Czerwonka L, De Santis RJ, Horowitz G et al. Staging cutaneous squamous cell carcinoma metastases to the parotid gland. Laryngoscope 127(9), 2063–2069 (2017).
    • 43. Estall V, Allen A, Webb A, Bressel M, Mccormack C, Spillane J. Outcomes following management of squamous cell carcinoma of the scalp: a retrospective series of 235 patients treated at the Peter MacCallum Cancer Centre. Australas. J. Dermatol. 58(4), e207–e215 (2017).
    • 44. Forest VI, Clark JJ, Veness MJ, Milross C. N1S3: a revised staging system for head and neck cutaneous squamous cell carcinoma with lymph node metastases – results of 2 Australian cancer centers. Cancer 116(5), 1298–1304 (2010).
    • 45. Goh RY, Bova R, Fogarty GB. Cutaneous squamous cell carcinoma metastatic to parotid – analysis of prognostic factors and treatment outcome. World J. Surg. Oncol. 10, 117 (2012).
    • 46. Gonzalez-Guerrero M, Martinez-Camblor P, Vivanco B et al. The adverse prognostic effect of tumor budding on the evolution of cutaneous head and neck squamous cell carcinoma. J. Am. Acad. Dermatol. 76(6), 1139–1145 (2017).
    • 47. Hinerman RW, Indelicato DJ, Amdur RJ et al. Cutaneous squamous cell carcinoma metastatic to parotid-area lymph nodes. Laryngoscope 118(11), 1989–1996 (2008).
    • 48. Hirshoren N, Danne J, Dixon BJ et al. Prognostic markers in metastatic cutaneous squamous cell carcinoma of the head and neck. Head Neck 39(4), 772–778 (2017).
    • 49. Kelder W, Ebrahimi A, Forest VI, Gao K, Murali R, Clark JR. Cutaneous head and neck squamous cell carcinoma with regional metastases: the prognostic importance of soft tissue metastases and extranodal spread. Ann. Surg. Oncol. 19(1), 274–279 (2012).
    • 50. Kosec A, Svetina L, Luksic I. Significance of clinical stage, extent of surgery and outcome in cutaneous squamous cell carcinoma of the head and neck. Int. J. Oral Maxillofac. Surg. 42(1), 82–88 (2013).
    • 51. Kraus DH, Carew JF, Horrison LB. Regional lymph node metastasis from cutaneous squamous cell carcinoma. Arch. Otolaryngol. Head Neck Surg. 124(5), 582–587 (1998).
    • 52. Li L, Tian Y, Shi C, Zhang H, Zhou Z. Over-expression of CD200 predicts poor prognosis in cutaneous squamous cell carcinoma. Med. Sci. Monit. 22, 1079–1084 (2016).
    • 53. Mcdowell LJ, Young RJ, Johnston ML et al. p16-positive lymph node metastases from cutaneous head and neck squamous cell carcinoma: no association with high-risk human papillomavirus or prognosis and implications for the workup of the unknown primary. Cancer 122(8), 1201–1208 (2016).
    • 54. Mclean T, Brunner M, Ebrahimi A et al. Concurrent primary and metastatic cutaneous head and neck squamous cell carcinoma: analysis of prognostic factors. Head Neck 35(8), 1144–1148 (2013).
    • 55. Mizrachi A, Hadar T, Rabinovics N et al. Prognostic significance of nodal ratio in cutaneous squamous cell carcinoma of the head and neck. Eur. Arch. Otorhinolaryngol. 270(2), 647–653 (2013).
    • 56. Oddone N, Morgan GJ, Palme CE et al. Metastatic cutaneous squamous cell carcinoma of the head and neck: the immunosuppression, treatment, extranodal spread and margin status (ITEM) prognostic score to predict outcome and the need to improve survival. Cancer 115(9), 1883–1891 (2009).
    • 57. Shao A, Wong DKC, Mcivor NP et al. Parotid metastatic disease from cutaneous squamous cell carcinoma: prognostic role of facial nerve sacrifice, lateral temporal bone resection, immune status and P-stage. Head Neck 36(4), 545–550 (2014).
    • 58. Sweeny L, Zimmerman T, Carroll WR, Schmalbach CE, Day KE, Rosenthal EL. Head and neck cutaneous squamous cell carcinoma requiring parotidectomy: prognostic indicators and treatment selection. Otolaryngol. Head Neck Surg. 150(4), 610–617 (2014).
    • 59. Tseros EA, Gebski V, Morgan GJ, Veness MJ. Prognostic significance of lymph node ratio in metastatic cutaneous squamous cell carcinoma of the head and neck. Ann. Surg. Oncol. 23(5), 1693–1698 (2016).
    • 60. Vasan K, Low TH, Gupta R et al. Lymph node ratio as a prognostic factor in metastatic cutaneous head and neck squamous cell carcinoma. Head Neck 23(10), 25066 (2018).
    • 61. Guyot P, Ades A, Ouwens MJ, Welton NJ. Enhanced secondary analysis of survival data: reconstructing the data from published Kaplan–Meier survival curves. BMC Med. Res. Methodol. 12(1), 9 (2012).
    • 62. Beasley GM, Kurtz J, Vandeusen J et al. Immune checkpoint inhibitor therapy as a novel and effective therapy for aggressive cutaneous squamous-cell carcinoma. Clin. Skin Cancer 1, 75–81 (2016).
    • 63. Berliner JG, Schulman JM, Lazarova Z, Olasz E, Arron ST. Response of cutaneous squamous cell carcinoma to treatment with cetuximab. Dermatol. Surg. 45(2), 313–316 (2019).
    • 64. Cavalieri S, Perrone F, Miceli R et al. Efficacy and safety of single-agent pan-human epidermal growth factor receptor (HER) inhibitor dacomitinib in locally advanced unresectable or metastatic skin squamous cell cancer. Eur. J. Cancer 97, 7–15 (2018).
    • 65. Cavalieri S, Perrone F, Milione M et al. PD-L1 expression in unresectable locally advanced or metastatic skin squamous cell carcinoma treated with anti-epidermal growth factor receptor agents. Oncology (Switzerland) 97(2), 112–118 (2019).
    • 66. Dereure O, Missan H, Girard C, Costes V, Guillot B. Efficacy and tolerance of cetuximab alone or combined with chemotherapy in locally advanced or metastatic cutaneous squamous cell carcinoma: an open study of 14 patients. Dermatology 232(6), 721–730 (2016).
    • 67. Di Monta G, Caraco C, Simeone E et al. Electrochemotherapy efficacy evaluation for treatment of locally advanced stage III cutaneous squamous cell carcinoma: a 22-cases retrospective analysis. J. Transl. Med. 15, 82 (2017).
    • 68. Ferrarotto R, Glisson B, Blumenschein G et al. Efficacy of pembrolizumab in patients with cutaneous squamous cell carcinoma. (2017). https://higherlogicdownload.s3.amazonaws.com/SITCANCER/3bcb5ebf-803a-42fe-83b6-0773bc4eb962/UploadedImages/Annual%20Meeting%202017/SITC_2017_Abstract_Book.pdf
    • 69. Foote MC, McGrath M, Guminski A et al. Phase II study of single-agent panitumumab in patients with incurable cutaneous squamous cell carcinoma. Ann. Oncol. 25(10), 2047–2052 (2014).
    • 70. Grob R, Gonzalez N, Basset-Seguin J et al. KEYNOTE-629: Phase II study of pembrolizumab for recurrent/metastatic or locally advanced unresectable cutaneous squamous cell carcinoma (CSCC). J. Clin. Oncol. 36(Suppl. 15), TPS9598 (2019).
    • 71. Guminski AD, Lim AML, Khushalani NI et al. Phase II study of cemiplimab, a human monoclonal anti-PD-1, in patients (pts) with metastatic cutaneous squamous cell carcinoma (MCSCC; group 1): 12-month follow-up. J. Clin. Oncol. 37, 9526 (2019).
    • 72. Guthrie TH Jr, Porubsky ES, Luxenberg MN, Shah KJ, Wurtz KL, Watson PR. Cisplatin-based chemotherapy in advanced basal and squamous cell carcinomas of the skin: results in 28 patients including 13 patients receiving multimodality therapy. J. Clin. Oncol. 8(2), 342–346 (1990).
    • 73. Hillen U, Leiter U, Haase S et al. Advanced cutaneous squamous cell carcinoma: a retrospective analysis of patient profiles and treatment patterns - results of a non-interventional study of the DeCOG. Eur. J. Cancer 96, 34–43 (2018).
    • 74. Huis In 't Veld EA Grunhagen DJ, Deroose JP et al. Isolated limb perfusion for unresectable extremity cutaneous squamous cell carcinoma; an effective limb saving strategy. Br. J. Cancer 119, 429–434 (2018).
    • 75. Kudchadkar RR, Yushak ML, Lawson DH et al. Phase II trial of pembrolizumab (MK-3475) in metastatic cutaneous squamous cell carcinoma (CSCC). J. Clin. Oncol. 36(Suppl. 15), 9543 (2018).
    • 76. Lippman SM, Parkinson DR, Itri LM et al. 13-cis-retinoic acid and interferon alpha-2a: effective combination therapy for advanced squamous cell carcinoma of the skin. J. Natl Cancer Inst. 84(4), 235–241 (1992).
    • 77. Maubec E, Boubaya M, Petrow P et al. Pembrolizumab as first line therapy in patients with unresectable squamous cell carcinoma of the skin: interim results of the Phase II CARSKIN trial. J. Clin. Oncol. 36(Suppl. 15), 9534 (2018). • Provides data for pembrolizumab in patients with CSCC.
    • 78. Migden MR, Khushalani NI, Chang ALS et al. Primary analysis of Phase II results of cemiplimab, a human monoclonal anti-PD-1, in patients (pts) with locally advanced cutaneous squamous cell carcinoma (laCSCC). J. Clin. Oncol. 37(Suppl. 15), 6015 (2019). • Provides practice-changing data that led to approval of cemiplimab for the treatment of advanced CSCC.
    • 79. Migden MR, Rischin D, Schmults CD et al. PD-1 blockade with cemiplimab in advanced cutaneous squamous-cell carcinoma. N. Engl. J. Med. 379, 341–351 (2018). • Provides practice-changing data that led to approval of cemiplimab for the treatment of advanced CSCC.
    • 80. Nottage MK, Lin C, Hughes BGM et al. Prospective study of definitive chemoradiation in locally or regionally advanced squamous cell carcinoma of the skin. Head Neck 39(4), 679–683 (2017).
    • 81. Owonikoko TK, Papadopoulos KP, Gil-Martin M et al. Phase I study of cemiplimab, a human monoclonal anti-PD-1, in patients with unresectable locally advanced or metastatic cutaneous squamous cell carcinoma (CSCC): longer follow-up efficacy and safety data. Ann. Oncol. 29(Suppl. 10), x25 (2018).
    • 82. Owonikoko TK, Papadopoulos KP, Johnson ML et al. Phase I study of cemiplimab, a human monoclonal anti-PD-1, in patients with unresectable locally advanced or metastatic cutaneous squamous cell carcinoma (CSCC): final efficacy and safety data. J. Clin. Oncol. 36(Suppl. 15), 9557 (2018).
    • 83. Papadopoulos KP, Johnson ML, Lockhart AC et al. First-in-human study of cemiplimab alone or in combination with radiotherapy and/or low dose cyclophosphamide in patients with advanced malignancies. Clin. Cancer Res. doi:10.1158/1078-0432.CCR-19-2609 (2019) (Epub ahead of print). • Provides safety, tolerability, dose-limiting toxicities, anti-tumor activity and pharmacokinetics of cemiplimab in patients with advanced solid tumors.
    • 84. Peyrade F, Kogay M, Viotti J et al. Cetuximab in patients with unresectable cutaneous squamous cell carcinoma is safe and effective: a real-life analysis. European Society for Medical Oncology Congress (2018). https://www.esmo.org/content/download/149891/2691140/file/ESMO-2018-Abstract-Book-partial-version.pdf
    • 85. Preneau S, Rio E, Brocard A et al. Efficacy of cetuximab in the treatment of squamous cell carcinoma. J. Dermatolog. Treat. 25(5), 424–427 (2014).
    • 86. Regeneron Pharmaceuticals, Inc./Sanofi. Individual patient data for the Phase I cemiplimab trial (R2810-ONC-1423).
    • 87. Regeneron Pharmaceuticals, Inc./Sanofi. Individual patient data for the Phase II cemiplimab trial (R2810-ONC-1540).
    • 88. Rischin D, Lim AM, Schmults CD et al. 1318P Phase II study of 2 dosing regimens of cemiplimab, a human monoclonal anti-PD-1, in metastatic cutaneous squamous cell carcinoma (mCSCC). Ann. Oncol. 30(Suppl. 5), mdz255.007 (2019).
    • 89. Rischin D, Migden MR, Chang A et al. Primary analysis of Phase II results for cemiplimab, a human monoclonal anti-PD-1, in patients with metastatic cutaneous squamous cell carcinoma (mCSCC). J. Clin. Oncol. 36(Suppl. 15), 9519 (2018).
    • 90. Rotunno R, Campana LG, Quaglino P et al. Electrochemotherapy of unresectable cutaneous tumours with reduced dosages of intravenous bleomycin: analysis of 57 patients from the International Network for Sharing Practices of Electrochemotherapy registry. J. Eur. Acad. Dermatol. Venereol. 32(7), 1147–1154 (2018).
    • 91. Sadek H, Azli N, Wendling JL et al. Treatment of advanced squamous cell carcinoma of the skin with cisplatin, 5-fluorouracil and bleomycin. Cancer 66(8), 1692–1696 (1990).
    • 92. Samstein RM, Ho AL, Lee NY, Barker CA. Locally advanced and unresectable cutaneous squamous cell carcinoma: outcomes of concurrent cetuximab and radiotherapy. J. Skin Cancer 2014, 284582 (2014).
    • 93. Yushak ML, Lawson DH, Goings M et al. Phase II trial of pembrolizumab (MK-3475) in metastatic cutaneous squamous cell carcinoma: an updated analysis. J. Clin. Oncol. 37(Suppl. 15), e21015 (2019).
    • 94. Bossi P, Cavalieri S, Perrone F et al. Efficacy and safety of single agent pan-HER inhibitor dacomitinib in locally advanced unresectable or metastatic skin squamous cell cancer (sSCC). J. Clin. Oncol. 35(Suppl. 15), 9543–9543 (2017).
    • 95. Jarkowski A 3rd, Hare R, Loud P et al. Systemic therapy in advanced cutaneous squamous cell carcinoma (CSCC): the Roswell Park Experience and a review of the literature. Am. J. Clin. Oncol. 39(6), 545–548 (2016). • Provides a review of the literature for patients with CSCC treated with systemic therapy.
    • 96. Maubec E, Boubaya M, Petrow P et al. Pembrolizumab as first-line therapy in patients with unresectable cutaneous squamous cell carcinoma (cSCC): Phase II results from CARSKIN. J. Clin. Oncol. 37(Suppl. 15), 9547–9547 (2019).
    • 97. Papadopoulos KP, Owonikoko TK, Johnson ML et al. REGN2810: a fully human anti-PD-1 monoclonal antibody, for patients with unresectable locally advanced or metastatic cutaneous squamous cell carcinoma (CSCC) – initial safety and efficacy from expansion cohorts (ECs) of Phase I study. J. Clin. Oncol. 35(Suppl. 15), 9503–9503 (2017).
    • 98. Nottage MK, Lin C, Hughes BG et al. Prospective study of definitive chemoradiation in locally or regionally advanced squamous cell carcinoma of the skin. Head Neck 39(4), 679–683 (2017).
    • 99. 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. 68(6), 957–966 (2013).