Loading

Review Article Open Access
Volume 4 | Issue 1 | DOI: https://doi.org/10.33696/cancerimmunol.4.058

Clinical, FDG-PET and Molecular Markers of Immune Checkpoint Inhibitor Response in Patients with Advanced Merkel Cell Carcinoma

  • 1Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria Australia
  • 2Department of Clinical Pathology and Centre for Cancer Research, University of Melbourne, Melbourne, Victoria, Australia
  • 3Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria Australia
  • 4Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria, Australia
+ Affiliations - Affiliations

*Corresponding Author

Shahneen Sandhu, shahneen.sandhu@petermac.org

Received Date: December 20, 2021

Accepted Date: January 11, 2022

Abstract

Recent findings: Merkel cell carcinoma (MCC) is a rare, highly aggressive, neuroendocrine cancer of the skin, associated with immunosuppression, Merkel cell polyoma virus (MCPyV) infection and UV-carcinogenesis. Whilst impressive and durable responses to immune checkpoint inhibitors have revolutionised the treatment of advanced MCC, approximately 50-70% of patients have either primary or acquired resistance to immune checkpoint blockade and robust predictive biomarkers of response are yet to be identified. Exploratory subgroup and biomarker analyses from clinical trials and retrospective studies have evaluated multiple clinical characteristics including age, performance status, immunosuppression, prior therapy, baseline and response imaging assessments, and molecular features including MCPyV status, programmed death ligand-1 (PD-L1) expression, tumour mutational burden (TMB) and tumour infiltrating lymphocytes as potential markers of response.

Summary: Better performance status, earlier line of therapy and early 18FDG-PET response have been consistently associated with favourable response to immune checkpoint inhibitors in MCC. High response rates are seen regardless of viral status, TMB and PD-L1 status.

Purpose of the review: We provide a review of clinical, imaging and molecular markers of response to immune checkpoint inhibitor therapy in MCC to aid patient selection and personalization of therapy.

Keywords

Merkel cell cancer, Immunotherapy, Immune checkpoint inhibitors, Biomarkers, FDG-PET, Merkel Cell Polyoma Virus, Tumour mutational burden, Programmed death ligand-1

Introduction

Merkel cell carcinoma (MCC) is a rare, highly aggressive, neuroendocrine cancer of the skin with an increasing incidence [1,2]. MCC occurs predominantly in older adults with risk factors including high exposure to ultra-violet (UV) light and immunosuppression [3]. The Merkel Cell Polyoma Virus (MCPyV) has been identified as a driver of a subset of MCC, with clonal integration of MCPyV DNA in MCC cells [4]. Abundant tumour infiltrating lymphocytes in a subset (~20%) [5-7] of MCPyV-positive tumours [8] and reactivity of T cells to viral-associated T-antigens [9,10] underpins the immunogenicity of MCPyV-positive MCC. In contrast, MCPyVnegative MCC are associated with UV signature and high tumour mutational burden (TMB), representing a second, distinct subtype with the potential to elicit anti-tumour immunity [11-13], as summarised in Box 1. MCPyV-mediated carcinogenesis predominates in the Northern hemisphere while UV-associated MCC are more common in regions of high UV exposure, including Australia [2,3,14-16].

Box 1: Merkel Cell Cancer Subtypes and Characteristics.
MCPyV-positive MCC MCPyV-negative MCC
Low TMB
Absence of UV signature Abundant TILs
Presence of MCPyV DNA and/or viral-associated antigens Predominates in North America and Europe
High TMB
Presence of UV signature
Absence of MCPyV DNA and viral associated antigens
More common in regions of high UV exposure (e.g. Australia)
MCPyV: Merkel Cell Polyoma Virus; MCC: Merkel Cell Cancer; UV signature: Ultra-Violet mutational signature; TILs: Tumour Infiltrating Lymphocytes

Prior to the advent of immunotherapy, prognosis for advanced MCC was dismal with a 5-year overall survival (OS) rate of 14-21% [17]. Although metastatic MCC is noted to be a chemotherapy-sensitive tumour, responses are shortlived with a median progression free survival (PFS) of 3 months [18]. Immune checkpoint inhibitors (ICI) have since revolutionised the treatment of advanced MCC. A number of ICI agents, including the anti-programmed death ligand 1 (anti-PD-L1) monoclonal antibody avelumab and anti-PD-1 monoclonal antibodies pembrolizumab and nivolumab have demonstrated impressive efficacy in advanced MCC, with 3-year survival rates approaching 60% [19]. Nevertheless, a proportion of patients either do not ever respond or develop resistance to ICI after initial response [20,21]. Key questions remain, including treatment strategies for primary non-responders and the optimal duration of ICI therapy in responders. Exploratory subgroup and biomarker analyses from clinical trials and retrospective studies can provide important insights into potential clinical, imaging and molecular markers of response to aid patient selection for ICI and personalization of treatment.

Immune Checkpoint Inhibition in MCC

ICI has been adopted as the mainstay of treatment for advanced MCC based on a number of phase II clinical trials. The landmark single-arm JAVELIN Merkel 200 study has established the anti-PD-L1 monoclonal antibody avelumab as the standard of care for the treatment of metastatic MCC. Part A of this study demonstrated an objective response rate (ORR) of 33% with impressive durability (median duration of response 40.5 months) in 88 patients that had progressed following chemotherapy [22,23]. In treatment-naïve patients enrolled in Part B of this study, the response rate was 62.1% [20]. Notably, 83% of responders experienced a duration of response (DOR) of at least 6 months, and 77.8% of responses were ongoing at the time of analysis [20]. A median overall survival of 20.3 months and 12.6 months has been reached for avelumab as first- and second-line treatment respectively [24,25], with a 12-month overall survival rate of 60% in the first-line (median follow up 21.2 months) [25] and a recently reported 5-year overall survival rate of 26% in the secondline setting (median follow up 65.1 months) [26]. In a realworld study of patients receiving first-line avelumab for metastatic MCC, the median overall survival was 20.2 months with an overall survival rate of 66.4% at 12 months [27]. The CITN-09/KEYNOTE-017 trial reported on the use of first-line pembrolizumab for unresectable stage III or metastatic MCC, with an ORR of 58% among 50 patients, of whom 86% had stage IV disease, and 14% had stage IIIB disease. The median DOR was not reached at three years, the median PFS was 16.8 months and 3-year overall survival rate was 59.4% in all comers and 89.5% in responders [28]. In the first-line advanced setting, 14.3 – 32% of patients demonstrate primary resistance to ICI [19,20]. In the KEYNOTE-017 trial, 11 of 29 (37.9%) patients with an initial partial or complete response later relapsed and 8 of these patients received subsequent immunotherapy, with treatment ongoing in 2 patients [19]. In the neoadjuvant setting, 2 doses of pre-operative nivolumab (240 mg, 2 weekly) resulted in a pathological complete response (pCR) rate of 47.2% in patients with resectable stage IIA – IV MCC in the CheckMate 358 trial [29]. The rarity of MCC has precluded large randomised controlled trials comparing ICI to chemotherapy. However, the unprecedented results of these single-arm studies, compared to historically poor outcomes, has cemented single-agent anti-PD-1 (pembrolizumab) and anti-PD-L1 (avelumab) as the front-line standard of care in metastatic MCC.

Clinical Markers of Response to Immune Checkpoint Inhibition in MCC

Clinical trials and retrospective case series have evaluated various clinical factors as potential markers of response. Key findings from these studies are summarised in Table 1. In keeping with the higher ORR observed in Part B (first-line, naïve to chemotherapy) compared to Part A (second-line, post-chemotherapy) of the JAVELIN Merkel 200 study, use of ICI as the first line of therapy has been consistently associated with favourable response, meeting statistical significance in the larger of two retrospective studies [13,30]. The basis of this differential response is poorly understood but may reflect an altered tumour immune environment and impaired adaptive immunity as a result of prior chemotherapy [31]. Better ECOG performance status also correlates with improved response in clinical trials [19] and case series [32]. Younger age has been associated with a trend towards improved response in some retrospective studies [30,32], although this has not been replicated in the clinical trial population [19]. In a retrospective series of 23 patients by Weppler et al., 6 of 6 patients who experienced an immune-related adverse event (irAE) responded (overall response 100% compared to 47% without irAE) [30]. These numbers are too small to meet statistical significance, however are supported by similar observations in patients with melanoma and NSCLC [33-35]. Conversely, immunosuppression was identified as a negative predictor of response in two retrospective studies [32,36]. Depth and duration of response appear to be prognostic: in the CheckMate-358 trial recurrence-free survival (RFS) significantly correlated with pCR with no relapses observed at 12 months of follow up in patients who achieved pCR [29]. In KEYNOTE-017, greater percentage of target lesion reduction and completion of 2 years of pembrolizumab were associated with improved overall survival at 30 months [19].

  Patients Clinical features Imaging features MCPyV status PD-L1/ PD-1 status TMB Other markers of response
ClinicalTrials
D’Angelo et al, JITC 2020; Kaufman
et al, JITC 2018;
D’Angelo et al ESMO Open 2021.
JAVELIN 200 Merkel Part A biomarker analysis and extended efficacy update
N = 88
1+priortherapyStage IV
<2 lines of therapy favours response (ORR 40.4% vs. 22.2%) NA MCPyV negative slightly favours response, not significant (ORR 35.5%
vs. 28.3%, NS)
PD-L1+ favours response (ORR 36.2%
vs 18.8%, NS)
Improved mOS in PD-L1+ patients (12.9m vs 7.3m)
TMB ≥ 2 NSSV/
Mb favours response (ORR 45.5% vs 28.0%),
6-month PFS and mOS (NR vs 12.6m)
GSEA: Interferon γ and interferon α and β pathways,
Th1, 2 and NK cell pathways enriched in responders
D’Angelo et al, JITC 2021.
JAVELIN200MerkelPart B updated overall survival and biomarker analysis
N = 116
No prior therapy Stage IV
NA NA MCPyV negative slightly favours response, not significant (ORR 48.6%
vs 34.2%, NS)
PD-L1 + favour response (ORR 61.9%
vs 33.3%, NS)
Improved mOS in PD-L1+ patients (NR vs 15.9m)
TMB > 2 NSSV/
Mb slightly favours response (ORR 50.0% vs 41.2%, NS), mOS (NR vs 17.2m)
Median or higher CD8+ T cell density at the invasive margin favours response (ORR 51.2% vs 28.6%, NS)
GSEA: interferon Interferon γ and interferon α and β pathways enriched in responders
Nghiem et al, NEJM, 2016.
CITN-09/ KEYNOTE-017
N = 26
No prior therapy Unresectable stage IIIB or IV
NA NA MCPyV positive favours response (ORR 62% vs. 44%) No correlation NA No correlation with CD8+ T cell infiltration and response
  N = 50
No prior therapy Unresectable stage IIIB or IV (86% stage IV)
Completion of 2 years          
  of therapy improves        
Nghiem et al, JITC 2021.
CITN-09/ KEYNOTE-017
3-year update and correlates
OS, HR 0.1 (95% CI
0.01-0.73)
ECOG 1 vs 0 reduced
survival, HR 2.70
(95%1.10-6.64)
CR/PR improves 3-year OS (89.5%
vs. 59.4%)
No difference in OS according to MCPyV status (HR 0.93, 95% CI
0.39-2.17)
No difference in OS (HR 0.48, 95% CI 0.19
-1.20)
NA
  No difference for        
  age, gender, baseline        
  tumour burden.        
Topalian et al, JCO 2020.
CheckMate 358
N = 39
Resectable stage IIA – IV
Neoadjuvant
12-month RFS 100% vs 59.6% for pCR vs non-pCR Imaging underestimates pCR
5 patients
with <30% radiographic reduction had pCR
No difference in response No difference in response No difference in TMB for pCR vs non-pCR observed Increased expression of CCL5, CXCL9, IL16, IL2RB in
responders (p < 0.05)
Retrospective Studies
      Low MTV at        
      baseline favours      
      response (p =      
    Age < 75 years favours 0.05)      
Weppler et al, JITC 2020. N = 23 response
(ORR 64% vs 50%, NS)
irAE favours response (ORR 100% vs 43%, NS)
CMR within 12 weeks correlates with improved survival
HR for PFS 0.31 (p = 0.38)
MCPyV negative favours response (ORR 69% vs 43%, NS) No correlation with response No correlation with response
      HR for OS 0.24      
      (p=0.19)      
Knepper et al, ACCR 2019. N = 317 Line of therapy favours response (ORR 75% 1L, 39% 2L, 18%
3L+, p = 0.006)
NA No difference in response
(ORR 50% vs 41%, p =
0.63)
PD-1+ favours response
(ORR 77% vs 21%, p
= 0.00598)
No difference in response
(ORR 50% vs
41%, p = 0.63)
 
Kacew et al, Oncotarget 2020. N = 45 Higher stage at primary disease diagnosis reduced odds of response (OR 0.06, p = 0.04)
Longer time to recurrence reduced odds of response (OR 0.75, p = 0.05)
NA No difference in response (p = 0.10)
No correlation with survival (p = 0.66)
NA No difference (median TMB
19.7 mut/Mb vs 4.8mut/MB, p = 0.11, for responders vs
non-responders)
SNVs in ARID2 and NTRK1 correlated with response (p=0.05)
Spassova et al, ACCR 2020. N = 41 ECOG 0 favours response
Absence of immunosuppression favours response
Age < 70 years favours response
NA No correlation with response No correlation with response NA Lower T cell clonality and higher TCR diversity seen in responders
Giraldo et al, JITC 2018. N = 26 NA NA NA No correlation with response for PD-L1 status using 1% as threshold
Higher PD-L1 correlated with response when analysed as a continuous variable (p = 0.02)
NA Higher density of PD-1+ cells in
responders vs non- responders (p = 0.03).
Density of PD-1+ cells adjacent to PD-L1+ correlates with response (p<0.05)

N: Number of patients; NA: Not Assessed; NS: Not Significant; NR: Not Reached; ORR: Objective Response Rate; Mos: Median Overall Survival; RFS: Recurrence Free Survival; OR: Odds Ratio; HR: Hazard Ratio; Pcr: Complete Pathological Response; CMR: Complete Response; PR: Partial Response; CMR: Completed Metabolic Response On FDG-PET; MTV: Metabolic Tumour Volume; 0L: Treatment naïve; 1L: 1 prior line of therapy; 2L: 2 prior lines of therapy; 3L+: 3 or more prior lines of therapy; SNVs: Single Nucleotide Variants; ECOG: Eastern Co-operative Oncology Group Performance Status (0-5); TCR: T cell Receptor; irAE: Immune-related Adverse Event; GSEA: Gene Set Enrichment Analysis.

Table 1: Summary of key factors associated with response in clinical trials and retrospective analyses.

It is unclear how prior or concurrent radiation impacts response to ICI. Radiation can induce immunogenic cell death and upregulate PD-L1 expression, potentially synergising with ICI [37,38]. One retrospective study of 45 patients found that patients with a longer disease-free interval between definitive radiation therapy and subsequent use of ICI for metastatic disease were less likely to respond (odds ratio 0.75, p =0.05). The authors hypothesised that more recent radiation in patients with shorter-disease free interval may improve response to ICI [36]. Weppler et al. described an overall response rate of 75% and complete response (CR) rate of 50% in four patients who received concurrent palliative radiation with commencement of ICI. Salvage radiation resulted in response in all 3 patients with isolated ICI-resistant lesions. However, all 3 patients who received salvage radiation in the setting of multisite progressive disease continued to experience disease progression at sites outside the radiation field [30].

Imaging Features of Response

The role for 2-[18F]fluoro-2-deoxy-D-glucose ([18F]FDG, 18FDG) positron emission tomography (PET) in assessing response and directing management in MCC was established in the pre-immunotherapy era [39,40]. A clear association between complete metabolic response (CMR) and improved overall survival in MCC has been demonstrated [41]. More recently, 18FDG-PET has been evaluated as a potential marker of response in patients treated with ICI. Patients who achieved CMR had a significantly lower metabolic tumour volume (MTV) at baseline [30], consistent with findings from clinical studies in MCC and melanoma that lower burden of disease is associated with favourable response to ICI [22,42]. These data support the role of routine surveillance 18FDG-PET to detect asymptomatic, smaller volume recurrence following definitive therapy for MCC [41]. Patients treated with ICI who achieved CMR on early 18FDG-PET (within 12 weeks of treatment initiation) demonstrated a trend towards improved progression free and overall survival [30]. These findings support the ongoing use of 18FDG-PET in response assessments and identify a need for specific research into additional treatment strategies for patients with high MTV.

Like other neuroendocrine tumours, approximately 70% of MCC express somatostatin receptors [43], presenting an opportunity for disease assessment with Gallium68 (68Ga)- labelled octreotide derivates using PET, the most widely used tracer being [68Ga]Ga-DOTA-octreotate (68Ga-DOTATATE). Several retrospective case series and retrospective analyses have demonstrated high sensitivity, specificity and diagnostic accuracy for 68Ga-DOTATATE PET in MCC [44]. Interestingly, when FDG-PET was directly compared to 68Ga-DOTATATE PET in MCC, similar avidity was seen with both tracers [45]. This is an unusual finding in poorly differentiated neuroendocrine tumours which are generally associated with lower SSTRexpression and higher FDG-avidity compared to their welldifferentiated counterparts, with prognostic implications [45]. There are no data on 68Ga-DOTATATE PET specifically assessing response to ICI, however this is being explored as the companion diagnostic for a potential theranostic strategy in metastatic MCC. The GOTHAM trial (NCT04261855) utilising 68Ga-DOTATATE PET to select patients to receive upfront 177Lu- DOTATATE (LuTate) peptide receptor radionuclide therapy in combination with avelumab.

Molecular Markers of Response

Despite the distinct molecular features of MCPyV-positive and MCPyV-negative MCC, both subtypes respond to immune checkpoint inhibition [13,21,23,29]. A clear predictive biomarker for response to ICI is yet to be elucidated. In addition to MCPyV status, tumour mutational burden (TMB) and PD-1 or PD-L1 status have been investigated as potential biomarkers in clinical trials. Changes in peripheral and intra-tumoral T cell populations and immune-related gene signatures have also been explored in retrospective studies.

MCPyV status

Traditionally, MCPyV-positive MCC has been associated with a more favourable prognosis [46], attributed to the increased immune-infiltrates and PD-L1 expression seen in this subtype [8,47]. Although responses to ICI have been observed in both MCPyV-positive and -negative cases of MCC, some differences in response rates have been observed. A recent real world retrospective study identified a non-significant trend towards improved ORR in MCPyV-negative tumours compared to MCPyV-positive tumours [30]. This is supported by a similar but non-significant finding in the JAVELIN Merkel 200 biomarker analysis with an ORR of 35.5% for MCPyV-negative MCC compared to 28.3% for MCPyV-positive MCC [23]. In contrast, a numerically higher ORR was seen in MCPyV-positive tumours in the initial analysis of the single-arm phase 2 trial of pembrolizumab (62% vs 44%) [21] although this finding was not reproduced in the subsequent 3-year update of this trial [19]. No association between MCPyV status and pathological response rate was seen in the neoadjuvant nivolumab trial [29], and a retrospective review of 57 patients with MCC by Knepper et al. did not find any difference in response according to MCPyV status [13]. Overall, no clearly consistent trend or statistically significant association between viral status and response to ICI has been observed.

Tumour mutational burden

High TMB is classically associated with high response rates to ICI across various tumour types [48]. Increased neoantigen expression is the putative mechanism of immunogenicity. Several studies have confirmed the bimodal distribution of TMB in MCC according to viral status, characteristically high in MCPyV-negative tumours and low in MCPyV-positive tumours, with very few (8%) intermediate cases [13,49]. In one large study of 317 patients, 37% were TMB high with a median TMB of 53.6 mutations per megabase (mut/Mb) and 55% were TMB-low with a median TMB 1.2 mut/Mb [13]. MCPyV DNA was not identified in any of the TMB high patients in this study [13], and was only detected at very low levels in another smaller study (median viral copy number 0.0037) [49]. Furthermore, 94% of patients with high TMB had a detectable UV mutational signature [13]. UV mutational signature and MCPyV DNA were mutually exclusive in intermediate TMB cases [13]. A clear molecular and oncogenic distinction between TMB-high, UVassociated and TMB-low, viral-driven MCC is thus drawn.

In the biomarker analyses of JAVELIN Merkel 200, patients with a TMB of 2 or more non-synonymous mutations per megabase had numerically but not significantly higher ORR compared to those with a lower TMB [23,25]. Despite this, no difference in the median TMB was seen between responders and non-responders was identified [25]. A non-significant trend towards improved PFS and median overall survival is noted in both first- and second-line settings (Table 1) [23,25]. None of these results met statistical significance and are limited by small population of only 36 evaluable patients. Interestingly, a very high ORR of 83.3% was seen in patients who exhibited high TMB in combination with another factor, such as high CD8+ T cell density at the invasive margin [23]. Contrary to these signals, no difference in TMB was observed between pathological responders and non-responders in the neoadjuvant nivolumab study, although only 14 patients were evaluable for TMB [29]. Multiple retrospective cohort studies have also failed to identify a clear correlation between TMB and response [13,30,36]. The absence of a definite association between high TMB and response in MCC differs from other tumour types and suggests an alternative mechanism of immunogenicity in MCPyV-positive tumours, presumably related to the presence of viral antigens and their ability to elicit an immune-response.

PD1/PDL1 status

PD-L1 status has been identified as a useful biomarker for response to ICI in some tumour types but not others. The JAVELIN 200 Merkel, KEYNOTE-017 and CheckMate 358 trials all included analyses of PD-L1 status. In the neoadjuvant nivolumab study, 27 patients had a quantifiable PD-L1 status: 7 had positive staining with greater than or equal to 1% of tumour cells staining for PD-L1 and 20 were deemed negative with staining less than 1% of tumour cells. No trends for radiographic or pathologic response or recurrence free survival according to PD-L1 status were observed [29]. Similarly, no correlation between PD-L1 status and response to CPI was observed in KEYNOTE-017 [21]. Although PD-L1 positive patients demonstrated a numerically higher ORR than PD-L1 negative patients in both first- and second-line settings in the JAVELIN Merkel 200 trial, this was not statistically significant [23,25] (Table 1). Long-term responses were observed in patients with both PD-L1 positive and negative tumours, although the majority (81.8%) of long-term survivors were PD-L1 positive [23]. A non-significant improvement in overall survival for PD-L1 positive patients is also seen in both cohorts (chemotherapy pretreated and chemotherapy naive) in JAVELIN Merkel 200 (Table 1) [25,26]. No significant correlation between PD-L1 status and response has been observed in retrospective studies [13,30].

Interestingly, one retrospective study also assessed PD-1 status on peritumoral lymphocytes and found a stark difference in response rate at 77% for PD-1 positive compared to 21% for PD-1 negative patients [13]. In an extended biomarker analysis of PD-1 and PD-L1 status from KEYNOTE-017, quantitative analysis showed higher densities of PD-1 and PD-L1 expression in responders compared to non-responders [50]. The geographic distribution of PD-1 and PD-L1 expressing cells also appeared to be meaningful, with higher responses seen in patients with a high number of PD-1 expressing immune cells adjacent to PD-L1 positive cells, indicative of adaptive rather than constitutive PD-L1 expression [50]. These findings suggest that categorical PDL1/ PD-1 positivity or negativity may be too simplistic as an approach to this biomarker. It is important to note that many of the studies evaluating PD-L1 status used different clones to assess PD-L1 expression, and whilst all considered tumours with 1% or greater staining as positive, some assessed tumour cells alone (tumour proportional score or TPS) [23,29,32] whereas others also included assessment of immune cells (IC score) [19,30], potentially contributing to the observed inconsistencies between studies.

T cells

Tumour infiltrating lymphocytes (TILs) have been shown to be prognostic in breast cancer and melanoma [51,52]. In MCC, T cell infiltration itself does not appear to be associated with response in several studies [19,23,30], although the updated biomarker analysis from the first-line cohort in JAVELIN indicates a trend towards improved ORR for patients with a median or higher density of CD8+ T cells at the invasive margin [25]. Additionally, some differences in T cell characteristics have been noted between responders and non-responders. Spassova et al. demonstrated lower clonality and more diverse T cell repertoire characterised the TILs of responders compared to non-responders [32]. Central memory T cells were the predominant TIL subtype in responders, a feature also associated with ICI efficacy in other tumour types including melanoma [32,53]. In another study evaluating peripheral mononuclear blood cells, the frequency of a highly activated CD8+ T cell subtype expressing both PD-1 and T cell immunoreceptor with Ig and ITIM domains (TIGIT) at baseline and following treatment with pembrolizumab was predictive of response [54]. These so-called “doublepositive” cells in peripheral blood samples are compelling as a potential biomarker and requires validation in larger patient populations.

Genes and gene sets

Several studies have identified enrichment of pathways involved in inflammation and immune response in responders to ICI [23,25,32]. Interferon gamma signatures were most common in responders, MCPyV-negative, PD-L1 positive tumours, and tumours with median or higher CD8+ T cell infiltration at the invasive margin [23,25]. Whilst MHC class I gene expression did not correlate with response or overall survival in the JAVELIN Merkel Part B biomarker analysis, increased MHC class I expression was associated with upregulation of inflammatory gene sets and was higher in patients with median or higher CD8+ T cell density [25]. Differential expression of genes involved in T cell attraction and activation has been noted in tumours of responders compared to non-responders [32]. While most emphasis has been placed on detection of conventional T cells, Gherardin et al. identified dominant infiltration of gamma delta T cells in some MCC and a gene-expression signature derived from these unconventional T cells was found elevated in bulk-tissue RNA from a subset of ICI-responsive cases [55]. Mutations in TP53 and RB1 are the most common genetic aberrations in both subtypes but are seen at higher frequencies in TMB-high population [13]. In a study of 45 patients, Kacew et al. found there were significantly more ARID2 and NTRK1 mutations in responders, a finding that requires further validation given small patient numbers [36].

Future Directions

Despite recent advances in the treatment of MCC, approximately 50 to 70 percent of patients demonstrate either primary or acquired resistance to immunotherapy within one year [20,22,28]. Definative predictive biomarkers of response to ICI are yet to be identified in MCC, with current research efforts limited by small patient numbers. Beyond further translational research to identify potential immune-related biomarkers and identify mechanisms of resistance, other approaches to be considered in the subset that exhibit primary and acquired resistance include treatment intensification with combined immunotherapy agents, targeted agents in conjunction with immunotherapy, or utilising multi-modality treatments, including radiation, to modulate the tumour microenvironment.

Combination ipilimumab and nivolumab has been explored in limited case series of patients resistant to anti-PD-L1 therapy, with response seen in 3 of the 5 treated patients [56,57]. This is a strong positive signal in a difficult to treat, refractory population, but requires validation with higher patient numbers. A randomised phase II trial of combination ipilimumab and nivolumab with or without radiation is Novel immuno-oncology agents including toll-like receptor ongoing, recruiting approximately 50 patients (NCT03071406). agonists [58] and T cell receptor therapy are being explored in MCC [59]. T cell receptor therapy is a focus of some early phase trials (NCT03747484) and involves the collection of T cells and programming high-affinity anti-MCPyV T-cell receptors into immature T cells for expansion.

Combining DNA Damage Repair (DDR) inhibitors, such as ATR (ataxia telangiectasia and Rad3-related) inhibitors, with ICI is another attractive strategy for MCC [60]. These agents target the cell cycle dysregulation that occurs as a result of TP53 and RB1 loss in MCC, and also induce immunogenic cell death, potentially augmenting the effect of ICI [60]. A number of ATR inhibitors have demonstrated tolerability in early-phase clinical trials [61-63] and several trials combining ATR inhibitors and ICI in other solid tumours are planned or ongoing (NCT05061134, NCT04216316, NCT04095273).

Trials to formally evaluate combining ICI with radiation with ICI in MCC are underway. In addition to the GOTHAM trial, NCT04261855 (evaluating avelumab with external beam radiotherapy and avelumab with LuTate in highly somatostatin receptor-expressing MCC in the metastatic setting), the CARTA trial (NCT04792073) is evaluating the role of comprehensive ablative radiation therapy with or without avelumab in patients with advanced disease progressing after anti-PD-1 therapy. In the adjuvant setting, the Immunotherapy Merkel Adjuvant Trial (I-MAT, NCT04291885) is randomising patients to receive concurrent, adjuvant avelumab or placebo following surgery and /or radiation for stage I-III MCC. Other ongoing adjuvant studies include the STAMP study (NCT03271372) and ADEMEC-O study (NCT02196961), randomising patients to observation or anti-PD-1 therapy (pembrolizumab and nivolumab respectively) after complete surgical resection of MCC, and the ADAM trial evaluating adjuvant avelumab following completion of surgery and/or radiotherapy (NCT03271372).

For patients who do respond to ICI, questions remain regarding the optimal duration of therapy. A retrospective study of 40 patients evaluated rates of progression following treatment cessation for response or otherwise and found relatively high rates of progression on treatment cessation, even among patients who initially achieved a CR [64]. Thirty (75%) patients were in CR at the time of treatment cessation and 26% of these patients and 57% of patients in partial response (PR) developed progression with a median time to progression of 5.5 months. Receiving fewer cycles of ICI was significantly associated with increased risk of progression. Ultimately, robust biomarkers are still required to identify those patients who will need intensified therapy, those who can be successfully treated with anti-PD-1/PD-L1 alone and those who can safely discontinue therapy after achieving a response.

Conclusion

Immune checkpoint inhibition has led to dramatic improvements for patients with advanced MCC. However, resistance remains a challenge and as yet there is no clear biomarker to aid patient selection or personalization of treatment. Performance status, line of therapy and early 18FDG-PET response have all been associated with favourable outcomes. Despite two clearly molecularly distinct subgroups of MCC, high ICI responses are seen in both subgroups, regardless of viral status, TMB and PD-L1 status. However, a significant subset of these patients who initially respond to ICI subsequently develop disease progression. More work is needed to identify and understand mechanisms of response and resistance to ICI, to select patients for standard or emerging therapies.

Disclosures

JDD, LSM, GK and AC have nothing to disclose. RWT has served on an advisory board for Merck Serono. SS has served on advisory board for Bristol Myer Squibb, Merck Sharp and Dohme, AstraZeneca, Janssen and has received grant funding to the institute from Pfizer, Merck Sharp and Dohme, AstraZeneca, Amgen, and Advanced Accelerators Applications (AAA), a Novartis Company (outside the submitted work). RJH holds shares in Telix Pharmaceuticals.

Author Contribution Statement

JDD, LM and SS devised the proof outline. All authors contributed to the final manuscript, with particular attention to respective areas of expertise from RJH and GK (nuclear medicine and imaging), and RWT and AC (immunology and molecular studies). SS provided oversight and supervision.

References

1. Paulson KG, Park SY, Vandeven NA, Lachance K, Thomas H, Chapuis AG, et al. Merkel cell carcinoma: current US incidence and projected increases based on changing demographics. Journal of the American Academy of Dermatology. 2018 Mar 1;8(3):457-63.

2. Youlden DR, Soyer HP, Youl PH, Fritschi L, Baade PD. Incidence and survival for Merkel cell carcinoma in Queensland, Australia, 1993- 2010. JAMA Dermatology. 2014 Aug 1;150(8):864-72.

3. Schadendorf D, Lebbé C, Zur Hausen A, Avril MF, Hariharan S, Bharmal M, et al. Merkel cell carcinoma: epidemiology, prognosis, therapy and unmet medical needs. European Journal of Cancer. 2017 Jan 1;71:53-69.

4. Feng H, Shuda M, Chang Y, Moore PS. Clonal integration of a polyomavirus in human Merkel cell carcinoma. Science. 2008 Feb 22;319(5866):1096-100.

5. Paulson KG, Iyer JG, Tegeder AR, Thibodeau R, Schelter J, Koba S, et al. Transcriptome-wide studies of merkel cell carcinoma and validation of intratumoral CD8+ lymphocyte invasion as an independent predictor of survival. Journal of Clinical Oncology. 2011 Apr 20;29(12):1539.

6. Sihto H, Böhling T, Kavola H, Koljonen V, Salmi M, Jalkanen S, et al. Tumor infiltrating immune cells and outcome of Merkel cell carcinoma: a population-based study. Clinical Cancer Research. 2012 May 15;18(10):2872-81.

7. Paulson KG, Iyer JG, Simonson WT, Blom A, Thibodeau RM, Schmidt M, et al. CD8+ lymphocyte intratumoral infiltration as a stage-independent predictor of Merkel cell carcinoma survival: a population-based study. American Journal of Clinical Pathology. 2014 Oct 1;142(4):452-8.

8. Lipson EJ, Vincent JG, Loyo M, Kagohara LT, Luber BS, Wang H, et al. PD-L1 expression in the Merkel cell carcinoma microenvironment: association with inflammation, Merkel cell polyomavirus, and overall survival. Cancer Immunology Research. 2013 Jul 1;1(1):54-63.

9. Iyer JG, Afanasiev OK, McClurkan C, Paulson K, Nagase K, Jing L, et al. Merkel cell polyomavirus-specific CD8+ and CD4+ T-cell responses identified in Merkel cell carcinomas and blood. Clinical Cancer Research. 2011 Nov 1;17(21):6671-80.

10. Longino NV, Yang J, Iyer JG, Ibrani D, Chow IT, Laing KJ, et al. Human CD4+ T Cells Specific for Merkel Cell Polyomavirus Localize to Merkel Cell Carcinomas and Target a Required Oncogenic Domain. Cancer Immunol. Res. 2019; 7: 1727–1739.

11. Harms PW, Vats P, Verhaegen ME, Robinson DR, Wu YM, Dhanasekaran SM, et al. The distinctive mutational spectra of polyomavirus-negative Merkel cell carcinoma. Cancer Research. 2015 Sep 15;75(18):3720-7.

12. Wong SQ, Waldeck K, Vergara IA, Schröder J, Madore J, Wilmott JS, et al. UV-associated mutations underlie the etiology of MCV-negative Merkel cell carcinomas. Cancer Research. 2015 Dec 15;75(24):5228-34.

13. Knepper TC, Montesion M, Russell JS, Sokol ES, Frampton GM, Miller VA, et al. The genomic landscape of Merkel cell carcinoma and clinicogenomic biomarkers of response to immune checkpoint inhibitor therapy. Clinical Cancer Research. 2019 Oct 1;25(19):5961-71.

14. Paik JY, Hall G, Clarkson A, Lee L, Toon C, Colebatch A, et al. Immunohistochemistry for Merkel cell polyomavirus is highly specific but not sensitive for the diagnosis of Merkel cell carcinoma in the Australian population. Human Pathology. 2011 Oct 1;42(10):1385-90.

15. Dabner M, McClure RJ, Harvey NT, Budgeon CA, Beer TW, Amanuel B, et al. Merkel cell Polyomavirus and p63 status in Merkel cell carcinoma by immunohistocnemistry: Merkel cell Polyomavirus positivity is inversely correlated with sun damage, but neither is correlated with outcome. Pathology. 2014 Apr 1;46(3):205-10.

16. Garneski KM, Warcola AH, Feng Q, Kiviat N, Leonard JH, Nghiem P. Merkel cell polyomavirus is more frequently present in North American than Australian Merkel cell carcinoma tumors. The Journal of Investigative Dermatology. 2009 Jan;129(1):246.

17. Harms KL, Healy MA, Nghiem P, Sober AJ, Johnson TM, Bichakjian CK, et al. Analysis of prognostic factors from 9387 Merkel cell carcinoma cases forms the basis for the new 8th edition AJCC staging system. Annals of Surgical Oncology. 2016 Oct;23(11):3564-71.

18. Becker JC, Lorenz E, Ugurel S, Eigentler TK, Kiecker F, Pföhler C, et al. Evaluation of real-world treatment outcomes in patients with distant metastatic Merkel cell carcinoma following second-line chemotherapy in Europe. Oncotarget. 2017 Oct 3;8(45):79731.

19. Nghiem P, Bhatia S, Lipson EJ, Sharfman WH, Kudchadkar RR, Brohl AS, et al. Three-year survival, correlates and salvage therapies in patients receiving first-line pembrolizumab for advanced Merkel cell carcinoma. Journal for Immunotherapy of Cancer. 2021;9(4).

20. D’Angelo SP, Russell J, Lebbé C, Chmielowski B, Gambichler T, Grob JJ, et al. Efficacy and safety of first-line avelumab treatment in patients with stage IV metastatic Merkel cell carcinoma: a preplanned interim analysis of a clinical trial. JAMA Oncology. 2018 Sep 1;4(9):e180077-.

21. Nghiem PT, Bhatia S, Lipson EJ, Kudchadkar RR, Miller NJ, Annamalai L,et al. PD-1 blockade with pembrolizumab in advanced Merkel-cell carcinoma. New England Journal of Medicine. 2016 Jun 30;374(26):2542-52.

22. Kaufman HL, Russell JS, Hamid O, Bhatia S, Terheyden P, D’Angelo SP, et al. Updated efficacy of avelumab in patients with previously treated metastatic Merkel cell carcinoma after≥ 1 year of follow-up: JAVELIN Merkel 200, a phase 2 clinical trial. Journal for Immunotherapy of Cancer. 2018 Dec;6(1):1-7.

23. D’Angelo SP, Bhatia S, Brohl AS, Hamid O, Mehnert JM, Terheyden P, et al. Avelumab in patients with previously treated metastatic Merkel cell carcinoma: long-term data and biomarker analyses from the single-arm phase 2 JAVELIN Merkel 200 trial. Journal for Immunotherapy of Cancer. 2020;8(1).

24. Nghiem P, Bhatia S, Brohl AS, Hamid O, Mehnert JM, Terheyden P, et al. Avelumab in patients with previously treated Merkel cell carcinoma (JAVELIN Merkel 200): Updated overall survival data after more than five years of follow up. J. Clin. Oncol.. 2021 May 20;39(15_ suppl):9517.

25. D’Angelo SP, Lebbé C, Mortier L, Brohl AS, Fazio N, Grob JJ, et al. First-line avelumab in a cohort of 116 patients with metastatic Merkel cell carcinoma (JAVELIN Merkel 200): primary and biomarker analyses of a phase II study. Journal for Immunotherapy of Cancer. 2021;9(7).

26. D’Angelo SP, Bhatia S, Brohl AS, Hamid O, Mehnert JM, Terheyden P, et al. Avelumab in patients with previously treated metastatic Merkel cell carcinoma (JAVELIN Merkel 200): updated overall survival data after> 5 years of follow-up. ESMO Open. 2021 Dec 1;6(6):100290.

27. Cowey CL, Liu FX, Kim R, Boyd M, Fulcher N, Krulewicz S, et al. Realworld clinical outcomes with first-line avelumab in locally advanced/ metastatic Merkel cell carcinoma in the USA: SPEAR-Merkel. Future Oncology. 2021 Jun;17(18):2339-50.

28. Nghiem P, Bhatia S, Lipson EJ, Sharfman WH, Kudchadkar RR, Brohl AS, et al. Three-year survival, correlates and salvage therapies in patients receiving first-line pembrolizumab for advanced Merkel cell carcinoma. Journal for Immunotherapy of cancer. 2021;9(4).

29. Topalian SL, Bhatia S, Amin A, Kudchadkar RR, Sharfman WH,Lebbé C, et al. Neoadjuvant nivolumab for patients with resectable Merkel cell carcinoma in the CheckMate 358 trial. Journal of Clinical Oncology. 2020 Aug 1;38(22):2476.

30. Weppler AM, Pattison A, Bhave P, De Ieso P, Raleigh J, Hatzimihalis A, et al. Clinical, FDG-PET and molecular markers of immune checkpoint inhibitor response in patients with metastatic Merkel cell carcinoma. Journal for Immunotherapy of Cancer. 2020;8(2).

31. Schadendorf D, Nghiem P, Bhatia S, Hauschild A, Saiag P, Mahnke L, et al. Immune evasion mechanisms and immune checkpoint inhibition in advanced merkel cell carcinoma. Oncoimmunology. 2017 Oct 3;6(10):e1338237.

32. Spassova I, Ugurel S, Terheyden P, Sucker A, Hassel JC, Ritter C, et al. Predominance of central memory T cells with high T-cell receptor repertoire diversity is associated with response to PD-1/PDL1 inhibition in Merkel cell carcinoma. Clinical Cancer Research. 2020 May 1;26(9):2257-67.

33. Maher VE, Fernandes LL, Weinstock C, Tang S, Agarwal S, Brave M, et al. Analysis of the association between adverse events and outcome in patients receiving a programmed death protein 1 or programmed death ligand 1 antibody. Journal of Clinical Oncology. 2019 Oct 20;37(30):2730-7.

34. Eggermont AM, Kicinski M, Blank CU, Mandala M, Long GV, Atkinson V, et al. Association between immune-related adverse events and recurrence-free survival among patients with stage III melanoma randomized to receive pembrolizumab or placebo: a secondary analysis of a randomized clinical trial. JAMA Oncology. 2020 Apr 1;6(4):519-27.

35. Akamatsu H, Murakami E, Oyanagi J, Shibaki R, Kaki T, Takase E, et al. Immune‐related adverse events by immune checkpoint inhibitors significantly predict durable efficacy even in responders with advanced non‐small cell lung cancer. The Oncologist. 2020 Apr;25(4):e679.

36. Kacew AJ, Dharaneeswaran H, Starrett GJ, Thakuria M, LeBoeuf NR, Silk AW, et al. Predictors of immunotherapy benefit in Merkel cell carcinoma. Oncotarget. 2020 Nov 24;11(47):4401.

37. Sharabi AB, Lim M, DeWeese TL, Drake CG. Radiation and checkpoint blockade immunotherapy: radiosensitisation and potential mechanisms of synergy. The Lancet Oncology. 2015 Oct 1;16(13):e498-509.

38. Gong J, Le TQ, Massarelli E, Hendifar AE, Tuli R. Radiation therapy and PD-1/PD-L1 blockade: the clinical development of an evolving anticancer combination. Journal for Immunotherapy of Cancer. 2018 Dec;6(1):1-7.

39. Ben-Haim S, Garkaby J, Primashvili N, Goshen E, Shapira R, Davidson T, et al. Metabolic assessment of Merkel cell carcinoma: the role of 18F-FDG PET/CT. Nuclear Medicine Communications. 2016 Aug 1;37(8):865-73.

40. George A, Girault S, Testard A, Delva R, Soulié P, Couturier OF, et al. The impact of 18F-FDG-PET/CT on Merkel cell carcinoma management: a retrospective study of 66 scans from a single institution. Nuclear Medicine Communications. 2014 Mar 1;35(3):282-90.

41. Byrne K, Siva S, Chait L, Callahan J, Bressel M, Seel M, et al. 15- Year experience of 18F-FDG PET imaging in response assessment and restaging after definitive treatment of Merkel cell carcinoma. Journal of Nuclear Medicine. 2015 Sep 1;56(9):1328-33.

42. Joseph RW, Elassaiss-Schaap J, Kefford R, Hwu WJ, Wolchok JD, Joshua AM, et al. Baseline tumor size is an independent prognostic factor for overall survival in patients with melanoma treated with pembrolizumab. Clinical Cancer Research. 2018 Oct 15;24(20):4960-7.

43. Gardair C, Samimi M, Touzé A, Coursaget P, Lorette G, Caille A, et al. Somatostatin receptors 2A and 5 are expressed in Merkel cell carcinoma with no association with disease severity. Neuroendocrinology. 2015;101(3):223-35.

44. Sachpekidis C, Sidiropoulou P, Hassel JC, Drakoulis N, Dimitrakopoulou-Strauss A. Positron emission tomography in Merkel cell carcinoma. Cancers. 2020 Oct;12(10):2897.

45. Taralli S, Sollini M, Milella M, Perotti G, Filice A, Menga M, et al. 18 F-FDG and 68 Ga-somatostatin analogs PET/CT in patients with Merkel cell carcinoma: a comparison study. EJNMMI Research. 2018 Dec;8(1):1-0.

46. Sihto H, Kukko H, Koljonen V, Sankila R, Böhling T, Joensuu H. Clinical factors associated with Merkel cell polyomavirus infection in Merkel cell carcinoma. JNCI: Journal of the National Cancer Institute. 2009 Jul 1;101(13):938-45.

47. Behr DS, Peitsch WK, Hametner C, Lasitschka F, Houben R, Schönhaar K, et al. Prognostic value of immune cell infiltration, tertiary lymphoid structures and PD-L1 expression in Merkel cell carcinomas. International Journal of Clinical and Experimental Pathology. 2014;7(11):7610.

48. Samstein RM, Lee CH, Shoushtari AN, Hellmann MD, Shen R, Janjigian YY, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nature Genetics. 2019 Feb;51(2):202-6.

49. Goh G, Walradt T, Markarov V, Blom A, Riaz N, Doumani R, et al . Mutational landscape of MCPyV-positive and MCPyV-negative Merkel cell carcinomas with implications for immunotherapy. Oncotarget. 2016 Jan 19;7(3):3403.

50. Giraldo NA, Nguyen P, Engle EL, Kaunitz GJ, Cottrell TR, Berry S, et al. Multidimensional, quantitative assessment of PD-1/PD-L1 expression in patients with Merkel cell carcinoma and association with response to pembrolizumab. Journal for Immunotherapy of Cancer. 2018 Dec;6(1):1-1.

51. Ali HR, Provenzano E, Dawson SJ, Blows FM, Liu B, Shah M, et al. Association between CD8+ T-cell infiltration and breast cancer survival in 12 439 patients. Annals of Oncology. 2014 Aug 1;25(8):1536-43.

52. Mihm Jr MC, Clemente CG, Cascinelli N. Tumor infiltrating lymphocytes in lymph node melanoma metastases: a histopathologic prognostic indicator and an expression of local immune response. Laboratory Investigation; a Journal of Technical Methods and Pathology. 1996 Jan 1;74(1):43-7.

53. Edwards J, Wilmott JS, Madore J, Gide TN, Quek C, Tasker A, et al. Cd103+ tumor-resident cd8+ t cells are associated with improved survival in immunotherapy-naïve melanoma patients and expand significantly during anti–pd-1 treatment. Clinical Cancer Research. 2018 Jul 1;24(13):3036-45.

54. Simon S, Voillet V, Vignard V, Wu Z, Dabrowski C, Jouand N, et al. PD-1 and TIGIT coexpression identifies a circulating CD8 T cell subset predictive of response to anti-PD-1 therapy. Journal for Immunotherapy of Cancer. 2020;8(2).

55. Gherardin NA, Waldeck K, Caneborg A, Martelotto LG, Balachander S, Zethoven M, et al. γδ T cells in Merkel cell carcinomas have a proinflammatory profile prognostic of patient survival. Cancer Immunology Research. 2021 Jun 1;9(6):612-23.

56. LoPiccolo J, Schollenberger MD, Dakhil S, Rosner S, Ali O, Sharfman WH, et al. Rescue therapy for patients with anti-PD-1- refractory Merkel cell carcinoma: a multicenter, retrospective case series. Journal for Immunotherapy of Cancer. 2019 Dec;7(1):1-5.

57. Glutsch V, Kneitz H, Gesierich A, Goebeler M, Haferkamp S, Becker JC, et al. Activity of ipilimumab plus nivolumab in avelumab-refractory Merkel cell carcinoma. Cancer Immunology, Immunotherapy. 2021 Jan 13:1-7.

58. Bhatia S, Miller NJ, Lu H, Longino NV, Ibrani D, Shinohara MM, et al. Intratumoral G100, a TLR4 agonist, induces antitumor immune responses and tumor regression in patients with Merkel cell carcinoma. Clinical Cancer Research. 2019 Feb 15;25(4):1185-95.

59. Gavvovidis I, Leisegang M, Willimsky G, Miller N, Nghiem P, Blankenstein T. Targeting Merkel cell carcinoma by engineered T cells specific to T-antigens of Merkel cell polyomavirus. Clinical Cancer Research. 2018 Aug 1;24(15):3644-55.

60. Goff PH, Bhakuni R, Pulliam T, Lee JH, Hall ET, Nghiem P. Intersection of Two Checkpoints: Could Inhibiting the DNA Damage Response Checkpoint Rescue Immune Checkpoint-Refractory Cancer?. Cancers. 2021 Jan;13(14):3415.

61. Yap TA, O’Carrigan B, Penney MS, Lim JS, Brown JS, de Miguel Luken MJ, et al. Phase I trial of first-in-class ATR inhibitor M6620 (VX- 970) as monotherapy or in combination with carboplatin in patients with advanced solid tumors. Journal of Clinical Oncology. 2020 Sep 20;38(27):3195.

62. Yap TA, O’Carrigan B, Penney MS, Lim JS, Brown JS, de Miguel Luken MJ, et al. Phase I trial of first-in-class ATR inhibitor M6620 (VX- 970) as monotherapy or in combination with carboplatin in patients with advanced solid tumors. Journal of Clinical Oncology. 2020 Sep 20;38(27):3195.

63. Kim ST, Smith SA, Mortimer P, Loembé AB, Cho H, Kim KM, et al. Phase I study of ceralasertib (AZD6738), a novel DNA damage repair agent, in combination with weekly paclitaxel in refractory cancer. Clinical Cancer Research. 2021 May 11.

64. Weppler AM, Da Meda L, Silva I, Xu W, Grignani G, Menzies AM, Carlino MS, Long GV, Nordman I, Steer C, Lyle M. Durability of response to immune checkpoint inhibitors (ICI) in metastatic Merkel cell carcinoma (mMCC) after treatment cessation. Journal of Clinical Oncology. 2021 39:9543-9543.

Author Information X