Skip to main content
Journal of Cancer Research and Clinical Oncology logoLink to Journal of Cancer Research and Clinical Oncology
. 2019 Jan 7;145(2):429–443. doi: 10.1007/s00432-019-02839-w

Immune checkpoint blockade for Merkel cell carcinoma: actual findings and unanswered questions

Marco Gallo 1,, Valentina Guarnotta 2, Federica De Cicco 3, Manila Rubino 4, Antongiulio Faggiano 3, Annamaria Colao 3; NIKE Group
PMCID: PMC11810318  PMID: 30617553

Abstract

Purpose

Merkel cell carcinoma (MCC) is a rare, aggressive neuroendocrine carcinoma arising from the skin. We aimed to review and deal with some of the most relevant controversial topics on the correct use of immunotherapy for the treatment of MCC.

Methods

The primary search was carried out via PubMed, EMBASE, and the Cochrane Library (until 31st May, 2018), while other articles and guidelines were retrieved from related papers or those referenced in these papers. Additionally, we performed an extensive search on ClinicalTrials.gov to gather information on the ongoing clinical trials related to this specific topic.

Results

We performed an up-to-date critical review taking into account the results of both retrospective and prospective published studies evaluating these issues: Are there any predictive criteria of response to immunotherapy? What is the correct place of immunotherapy in the treatment algorithm of MCC? What is the best choice after immunotherapy failure? What to do with patients for whom immunotherapy is not been feasible or contraindicated? How long should immunotherapy be prolonged, and what follow-up should be offered after complete response?

Conclusion

The therapeutic landscape of MCC is rapidly evolving: many open issues will probably be resolved, and many other questions are likely to arise in the next few years. The results of ongoing prospective clinical trials and of several other studies on these issues are eagerly awaited.

Keywords: Merkel cell carcinoma, Neuroendocrine tumours, Immune checkpoint inhibitors, Avelumab, Pembrolizumab, Therapy

Introduction

Merkel cell carcinoma (MCC) is an uncommon, aggressive neuroendocrine carcinoma arising in the dermoepidermal junction of the skin, that was originally described less than 50 years ago (Toker 1972). MCC, which is the second most common cause of skin cancer death after melanoma, has been traditionally believed to arise from skin mechanoreceptors with touch-sensitivity function (Merkel cells, as named after the German scientist who discovered them in 1875), but its histogenesis is still controversial. According to another hypothesis, MCC could originate from an immature totipotential stem cell with neuroendocrine features acquired during malignant transformation (Tilling and Moll 2012).

The estimated annual incidence of MCC in 2006 was 0.6 per 100,000 person (Albores-Saavedra et al. 2010), with higher incidence rates among older males and subjects with light skin. However, a younger age and higher incidences are observed in immunocompromised people, such as organ transplant recipients, HIV-infected subjects, people using immunosuppressant medications, and those with lymphoproliferative disorders or other malignancies (Tadmor et al. 2011; Engels et al. 2002; Clarke et al. 2015). The main factors associated with the pathogenesis of MCC include infection with the Merkel cell polyomavirus (MCPyV), ultraviolet radiation exposure (UVB irradiation), and immunosuppression. The incidence of MCC is increasing more rapidly than other skin tumours, such as malignant melanoma, probably due to longevity, improved detection, and increased reporting (Hodgson 2005; Prewett and Ajithkumar 2015).

Most frequently, MCC presents with local disease, but regional lymph node and distant metastases may be present in up to 30% of new cases. In a minority of cases, MCC is diagnosed as a lymph node metastasis without an identifiable primary lesion, which may have spontaneously regressed or be occult (Deneve et al. 2012). Typically, patients with occult primary MCCs have higher tumour mutational burden, lower association with MCPyV, and better prognosis than those with known primary, possibly due to an effective immune response and immunologically mediated regression (Uchi 2018).

Even though several clinicopathologic features may have an influence on prognosis (e.g. increasing age, male sex, histological features, MCPyV serology, and immune status), the involvement of regional lymph nodes or distant metastases at presentation are the most important predictors of survival for MCC. In 2017, a new MCC-specific consensus staging system has been developed by the American Joint Committee on Cancer (AJCC), taking into account size of the primary lesion and presence of tumour invasion (T), presence of nodal metastasis (N), and distant metastasis (M) (Bichakjian et al. 2017). Based upon TNM information, patients with MCC are assigned to prognostic stage groups: stages I and II MCC include patients with local disease only (≤ 2 and > 2 cm maximum tumour dimension, respectively), stage III includes patients with regional nodal disease, and stage IV is for patients with metastases beyond the regional lymph nodes.

The treatment depends on the stage of the disease, the tumour site and any comorbid conditions. Excision of MCC with 1- to 2-cm negative margins (with sentinel lymph node biopsy for patients with clinically negative nodes) is the mainstay of therapy for local disease (stage I and II), followed by adjuvant radiotherapy. The latter can be omitted with small tumours (≤ 1 cm), wide excision margins (> 2 cm), negative lymph nodes, and no other adverse features (such as immunosuppression).

Positive nodal disease (stage III), affecting one-third of patients, should be treated with neck dissection and adjuvant radiotherapy. Radiotherapy (45–50 Gy for microscopic disease and 50–60 Gy for macroscopic disease) is an option for primary therapy in patients who cannot undergo or not amenable to surgical interventions.

Until recently, guidelines suggested chemotherapy in cases of metastatic, advanced-stage MCC (stage IV), however, recommending enrolment in clinical trials due to the poor long-term outcomes of chemotherapy. Systemic chemotherapy with therapeutic regimes derived from small-cell lung cancer due to the similar neuroendocrine properties to MCC, and has been typically performed with platinum, etoposide, doxorubicin, cyclophosphamide, vincristine, paclitaxel, and topotecan, alone or in combinations. Albeit high response rates (from 52 to 61%), response duration is usually short (3–15 months) and survival benefits questionable (Prewett and Ajithkumar 2015; Hasan et al. 2013). The few real-world, retrospective studies which assessed second-line or later chemotherapy showed low objective response rates (ORRs; from 8.8 to 23.0% with no complete responses) and very limited durability (1.3–3.3 months) (Iyer et al. 2016; Becker et al. 2017). Furthermore, most regimens are associated with considerable toxicity and can worse immunosuppression, paradoxically stimulating the tumour (Nghiem et al. 2017; Rabinowitz 2017). Indeed, considerable evidence suggests that immunosuppression contributes significantly to the course and prognosis of MCC, implying that therapeutic agents that promote antitumour immune responses might be beneficial in MCC.

One mechanism contributing to tumour proliferation is the expression of programmed cell death ligand 1 (PD-L1, an immune-inhibitory ligand) in the tumour microenvironment. PD-L1 is an immune checkpoint molecule that binds to its main receptor, PD-1, which is expressed by activated T lymphocytes. The complex PD-L1/PD-1 inhibits the signalling pathways involved in T-cell proliferation, survival and cytotoxic activity (including cytokine and enzymes release), preventing overstimulation of immune responses. Tumour associated inflammation has been shown to upregulate PD-L1 expression, thus enabling tumours to avoid and escape immune surveillance. Therefore, blocking the interaction between PD-1 and PD-L1 is a key therapeutic target in the reactivation of the immune response and a promising target for many tumours, including MCC. Indeed, PD-L1 expression has also been described in MCC cells and in tumour-infiltrating and peritumoural leukocytes, providing a rationale for treatment with immune checkpoint inhibitors (ICIs) in MCC (Nghiem et al. 2017). Both avelumab (MSB0010718C; anti-PD-L1) and pembrolizumab (anti-PD-1) have recently shown promising results in clinical trials performed on patients with metastatic MCC, as discussed further in the manuscript. Results of these trials led to the addition of ICIs in the most recent update of the National Comprehensive Cancer Network (NCCN) Clinical Practice guidelines as a treatment option for stages III–IV MCC (Bichakjian et al. 2018). Since 2017, based on its therapeutic responses and safety profile, avelumab has been approved in the USA, the European Union, Switzerland, and Japan for the treatment of metastatic MCC, in first and second-line settings. Furthermore, on December 2018, the US Food and Drug Administration granted accelerated approval to pembrolizumab for adult and paediatric patients with recurrent locally advanced or metastatic MCC. Most likely, chemotherapy will no longer represent the first option for advanced stage MCC.

Recent prospective clinical trials in patients with advanced MCC revolutionised medical therapy of stage IV MCC, showing the durable efficacy and favourable tolerability of anti–PD-1 or anti–PD-L1 ICIs in the first-line or second-line and later settings.

However, many critical issues on the correct use of ICIs for the treatment of MCC still remain and many questions are still unanswered (Table 1).

Table 1.

Controversial issues about immunotherapy for MCC

Are there any predictive criteria of response to immunotherapy? (i.e. can patients be selected for a better response to immunotherapy?)

What is the place of immunotherapy in the treatment algorithm of Merkel cell carcinoma?

Can immunotherapy be helpful as first-line therapy for stage IV disease, or it should be used only after PD with systemic chemotherapy?

Can immunotherapy be an option as an adjuvant treatment for regional disease (stage III MCC)?

Is there a role for neoadjuvant therapy with immune checkpoint inhibitors?

What are the advantages and the potential drawbacks of the association of immunotherapy and radiotherapy?

What are the options for patients who progress after immunotherapy?

Patients who not respond to immunotherapy

Patients who develop resistance or escape to immunotherapy

What are the options for patients who are not candidates for immunotherapy?

Patient categories who were excluded from clinical trials

Patients with active autoimmune disorders

Patients who underwent organ or bone marrow transplantation

Patients with abnormal organ and bone marrow function

Patients with immunodeficiency (e.g. HIV) or ongoing systematic immunosuppressive therapy

How long should treatment with immune checkpoint inhibitors be prolonged after complete response?
What follow-up should be performed after complete response?

With the aim to review and deal with some of the most relevant controversial topics on the management of MCC with ICIs, we performed an up-to-date critical review taking into account the results of both retrospective and prospective published studies. The primary search was carried out via PubMed, EMBASE, and the Cochrane Library (until 31st May, 2018), while other articles and guidelines were retrieved from related papers or those referenced in these papers. The search was restricted to reports published in English. Additionally, we performed an extensive search on ClinicalTrials.gov to gather information on the ongoing clinical trials related to this specific topic.

Are there any predictive criteria of response to immunotherapy?

Viral antigens of virus-positive MCC can act as excellent immune targets. On the other hand, virus-negative MCC express a higher load of somatic mutations and putative tumour neoantigens, probably induced by UV irradiation (Goh et al. 2016; Amaral et al. 2017). Therefore, both virus-positive and virus-negative MCC express non-self antigens making these tumours excellent candidates for immunotherapy (Goh et al. 2016; Amaral et al. 2017; Baker et al. 2018). However, since checkpoint-based immunotherapy is not effective for all MCC patients, it would be of the uttermost importance to better understand the cellular and genomic determinants of the immune response in this setting, to better select candidate patients for immunotherapy.

Indeed, durable responses to therapies involving the PD-1/PD-L1 axis have recently been shown to occur irrespectively of tumour MCPyV status, as well as both in patients with PD-L1 + and with PD-L1- tumours (Kaufman et al. 2016; Nghiem et al. 2016). This may be due to the relative small number of patients treated in the two phase II studies performed with avelumab and pembrolizumab. As an example, PD-L1 expression in the primary tumour and/or in the metastatic lesions is heterogeneous, thus a negative single biopsy may not be representative of all the tumours (Becker et al. 2018).

Other characteristics of the tumour microenvironment could also modulate the immune response correlating with clinical outcomes of anti-PD-1 or anti-PD-L1 therapy, such as the presence of CD8 + T-cell infiltrates, the expression of additional immune checkpoints, and the gene-expression profile (Schadendorf et al. 2017; Becker et al. 2018).

Ongoing research in MCC is trying to identify reliable biomarkers that will help predict which patients have more chances to respond to immunotherapy. Implementing immunotherapy for MCC might require, in the future, precision medicine approaches for any given case (Cassler et al. 2016). Actually, however, no powerful predictor of response can be used to decide whether to use immunotherapy in a patient with MCC (Hauschild and Schadendorf 2016; Bommareddy and Kaufman 2017).

As of today, immunotherapy should be considered for first-line treatment in patients with advanced MCC, since a trend toward a higher response rate has been observed in patients with fewer lines of prior treatment (D’Angelo et al. 2018). Furthermore, previous chemotherapy has recently shown to decrease the expression of both PD-L1 and PD-1 in different settings (Rojkó et al. 2018).

What place for immunotherapy in the treatment algorithm of Merkel cell carcinoma?

Immunotherapy is currently recommended for patients with disseminated disease or recurrence. Data from prospective clinical trials showed that the rates of durable responses are improved with immunotherapy than cytotoxic treatment, with minor side effects both in the first and second/later line of therapy (Kaufman et al. 2016; Nghiem et al. 2016) and a better cost-effectiveness (Bharmal et al. unpublished results).

In a multicenter Phase II trial of 88 patients with stage IV chemotherapy refractory MCC, treatment with avelumab showed durable responses: the ORR was 33% (95% CI 23–44), with 74% of responses lasting ≥ 1 year and a 1-year overall survival (OS) rate of 52% (95% CI 41–62) in second-line and further-line therapies (Kaufman et al. 2016, 2018). In addition, a trend towards a higher response rate was observed in those patients with fewer lines of previous chemotherapy. Preliminary data have been recently published on the favourable effects of avelumab in treatment naïve patients with metastatic MCC, showing an ORR of 62.1% and a CR of 13.8% at 6 months (D’Angelo et al. 2018). Interestingly, a cross-study analysis of avelumab after ≥ 1 line of chemotherapy in metastatic MCC after ≥ 1 year of follow-up was performed, finding that second-line chemotherapy was associated with a lower ORR than avelumab (9–23% vs. 33%, respectively), and with a 1-year progression-free survival (PFS) rate of 0% vs. 30%, respectively (Kaufman et al. 2018). Data from a large cohort of patients, enrolled in a global expanded access program and treated with avelumab as second-line treatment in a real world setting, showed an ORR (as evaluated by the treating physician) of 52.5% in the global population and 54.8% in the European population (Nathan et al. unpublished results).

In another multicenter, phase II non-controlled study in patients with advanced (stage III or IV) MCC, pembrolizumab administered as a first-line treatment showed an ORR of 56% (95% CI 35–76), with 86% of responses ongoing at data cutoff (Nghiem et al. 2016). Interestingly, in the expanded phase of this study ORR was 50% (95% CI 34.2–65.8; CR 19%, PR 31%) in 42 patients with ≥ 21 week of follow-up. OS rate at 18 months was 68% and median OS was not reached. Among 21 confirmed responders, median response duration was not reached (range 3.9–25.6 months) (Nghiem et al. 2018).

Other immunotherapies being investigated in MCC include ipilimumab and nivolumab (anti-PD-1) as first- or second-line treatment of unresectable local and/or metastatic MCC (stages II–IV) (Topalian et al. 2018). As an example, preliminary results on nivolumab as first- or second-line treatment, in patients with advanced MCC, showed an ORR of 68% and a PFS at 3 months of 82% (ongoing trial NCT02488759).

Collectively, these data suggest that immunotherapies can substantially improve the outcome of patients with metastatic MCC. Therefore, avelumab, nivolumab and pembrolizumab are currently recommended for patients with advanced MCC.

Immunotherapy as an adjuvant treatment for regional disease (stage III)

The possibility to use immunotherapy in the adjuvant setting following surgical resection of local MCC seems appropriate, due to the consideration that an early treatment of a tumour correlates with an improved prognosis and to the successful approach in other tumours such as melanoma (Eggermont et al. 2015, 2018; Gibney et al. 2015; Weber et al. 2017).

Currently, the efficacy of avelumab as adjuvant therapy versus placebo in patients with stage III/IIIb MCC, with or without radiotherapy, is under investigation (NCT03271372). A clinical trial on nivolumab and ipilimumab in the adjuvant setting (NCT02196961) is partially ongoing, too (ADMEC-O) (See Table 2). In the arm of this study evaluating ipilimumab in 40 patients within 12 weeks after complete tumour resection, no significant differences were observed between ipilimumab and observation (hazard ratio 1.8; 95% CI 0.3–10; p = 0.48) (Becker et al. unpublished results). However, patients in the treatment arm had a significantly increased incidence of adverse events. For these reasons, the enrolment in this arm was stopped.

Table 2.

Ongoing active clinical trials for MCC.

(source: https://www.clinicaltrials.gov)

NCT Title Phase Intervention
NCT02196961 Adjuvant Therapy of Completely Resected Merkel Cell Carcinoma With Immune Checkpoint Blocking Antibodies versus Observation (ADMEC-O) II

Ipilimumab

Nivolumab

NCT02488759 Non-Comparative, Open-Label, Multiple Cohort, Phase 1/2 Study of Nivolumab Monotherapy and Nivolumab Combination Therapy in Subjects With Virus-Positive and Virus-Negative Solid Tumors

I

II

Nivolumab

Ipilimumab

BMS-986016 Daratumumab

NCT03167164 NANT Merkel Cell Carcinoma (MCC) Vaccine: Combination Immunotherapy in Subjects With MCC Who Have Progressed on or After Anti-programmed Death-ligand 1 (PD-L1) Therapy

I

II

Avelumab

Bevacizumab

Capecitabine

Cisplatin

Cyclophosphamide

5-fluorouracil

Leucovorin

Nab-Paclitaxel

Omega-3-acid ethyl esters

Stereotactic Body Radiation Therapy

+ 5 biological

NCT02465957 Phase II Study of aNK (Activated NK-92 Natural Killer Cells) Infusions in Combination With ALT-803 (IL-15) in Patients With Stage III (IIIB) or Stage IV Merkel Cell Carcinoma (MCC) II aNK (NK-92)
NCT02036476 Cabozantinib in Recurrent/Metastatic Merkel Cell Carcinoma II Cabozantinib
NCT03071406 A Phase II, Randomized, Multi-institutional Study of Nivolumab and Ipilimumab versus Nivolumab, Ipilimumab and Stereotactic Body Radiation Therapy for Metastatic Merkel Cell Carcinoma II

Nivolumab

Ipilimumab

Stereotactic Body Radiation Therapy

NCT03271372 A Multicenter, Randomized, Double-Blinded, Placebo-Controlled, Phase III Trial of Adjuvant Avelumab (Anti-PDL-1 Antibody) in Merkel Cell Carcinoma Patients With Clinically Detected Lymph Node Metastases III

Avelumab

Placebo

NCT02155647 A Phase II, Open-Label, Multicenter Trial to Investigate the Clinical Activity and Safety of Avelumab (MSB0010718C) in Subjects With Merkel Cell Carcinoma II Avelumab
NCT01013779 A Phase II Efficacy Study of Chemo-Radiotherapy in PET Stage II and III Merkel Cell Carcinoma of the Skin II Carboplatin Etoposide Radiotherapy
NCT02584829 Study to Evaluate Cellular Adoptive Immunotherapy Using Polyclonal Autologous CD8 + Antigen-Specific T Cells for Metastatic Merkel Cell Carcinoma in Combination With MHC Class I Up-Regulation and the Anti-PD-L1 Antibody Avelumab

I

II

Avelumab

MCPyV TAg-specific Polyclonal Autologous CD8-positive T Cells Radiation Therapy Recombinant Interferon Beta

NCT02819843 A Phase II Randomized Trial of Intralesional Talimogene Laherparepvec (TALIMOGENE LAHERPAREPVEC) With or Without Radiotherapy for Cutaneous Melanoma, Merkel Cell Carcinoma, or Other Solid Tumors II TALIMOGENE LAHERPAREPVEC (TVEC) Hypofractionated Radiotherapy
NCT03304639 A Randomized Phase II Study of Anti-PD1 Antibody [MK-3475 (Pembrolizumab)] Alone versus Anti-PD1 Antibody Plus Stereotactic Body Radiation Therapy in Advanced Merkel Cell Carcinoma II Pembrolizumab Stereotactic Body Radiation Therapy
NCT02267603 A Phase II Study of MK-3475 in Patients With Advanced Merkel Cell Carcinoma (MCC) II Pembrolizumab
NCT02978625 A Phase II Study of Talimogene Laherparepvec Followed by Talimogene Laherparepvec + Nivolumab in Refractory T Cell and NK Cell Lymphomas, Cutaneous Squamous Cell Carcinoma, Merkel Cell Carcinoma, and Other Rare Skin Tumors II Nivolumab Talimogene Laherparepvec
NCT03458117 A Phase I, Open Label, Single Arm, Single Centre Study to Evaluate Mechanism of Action of Talimogene Laherparepvec (T-VEC) in Locally Advanced Non-melanoma Skin Cancer I Talimogene Laherparepvec (T-VEC)
NCT03435640 A Phase 1/2, Open-label, Multicenter, Dose Escalation and Dose Expansion Study of NKTR-262 in Combination With NKTR-214 and in Combination With NKTR-214 Plus Nivolumab in Patients With Locally Advanced or Metastatic Solid Tumor Malignancies

I

II

NKTR-262

NKTR-214 Nivolumab

NCT02890368 A Phase 1 Dose Escalation Trial of Intratumoral Injections of TTI-621 in Subjects With Relapsed and Refractory Percutaneously-Accessible Solid Tumors and Mycosis Fungoides I

TTI-621

TTI-621 + PD-1/PD-L1 inhibitor

TTI-621 + pegylated interferon-α2a

TTI-621 + T-Vec

TTI-621 + radiation

NCT02643303 A Phase 1/2 Study of In Situ Vaccination With Tremelimumab and IV Durvalumab (MEDI4736) Plus the Toll-like Receptor Agonist PolyICLC in Subjects With Advanced, Measurable, Biopsy-accessible Cancers

I

II

Durvalumab

Tremelimumab

Poly ICLC

NCT03212404 A Phase 1, Open-label, Multicenter, Dose-escalation Study of CK-301 Administered Intravenously as a Single Agent to Subjects With Advanced Cancers I CK-301
NCT03241173 A Phase 1/2 Study Exploring the Safety, Tolerability, and Efficacy of INCAGN01949 in Combination With Immune Therapies in Subjects With Advanced or Metastatic Malignancies

I

II

INCAGN01949

Nivolumab

Ipilimumab

NCT03071757 A Multicenter, Phase 1, Open-Label, Dose-Escalation Study of the Safety, Tolerability and Pharmacokinetics of ABBV-368 as a Single Agent and Combination in Subjects With Locally Advanced or Metastatic Solid Tumors I ABBV-368 Nivolumab
NCT03000257 A Multicenter, Phase 1, Open-Label, Dose-Escalation Study of ABBV-181, a Monoclonal Antibody, as Monotherapy and in Combination With Another Anti-Cancer Therapy in Subjects With Advanced Solid Tumors I

Rovalpituzumab Tesirine

ABBV-181

Therefore, the use of PD-1/PD-L1 inhibitors in the adjuvant setting has been a major discussion topic in the last years. Some of the challenges of assessing these drugs in the adjuvant setting include the identification of which patients may have the best benefit from it, without exposing to unnecessary, ineffective, and toxic treatment (Napolitano et al. 2018).

Immunotherapy as a neoadjuvant therapy

The use of ICIs as a neoadjuvant treatment, before surgical resection, has been investigated, resulting advantageous in mice models and in humans with non-small-cell lung carcinoma (NSCLC). The mechanism of action seems to be the generation of circulating T cells, which may improve protection against tumour growth (Liu et al. 2016; Forde et al. 2018). Neoadjuvant ipilimumab treatment showed favourable effects in patients with regionally advanced melanoma (Tarhini et al. 2014). Currently, a study evaluating the combination of neoadjuvant ipilimumab and nivolumab in patients with melanoma is ongoing (NCT02977052), while no data are available for patients with MCC.

Immunotherapy ± radiotherapy

The combination of immunotherapy and radiotherapy has been shown to exert favourable effects on tumour control, due to a synergistic effect (Sharabi et al. 2015).

Indeed, radiotherapy has been demonstrated to stimulate T-cell activation and proliferation by releasing tumour antigens, and to modulate the expression of immune checkpoint ligands, including PD-L1, on tumour cells (Parker et al. 2013; Deng et al. 2014). In addition, it is correlated to an abscopal effect, a phenomenon in which tumour regression occurs in non-irradiated lesions (Demaria et al. 2004). For these reasons, sequential use of radiotherapy and ICI may be associated with an improvement of relapse free survival.

Available data on NSCLC and metastatic melanoma have shown clinical benefit when immunotherapy was given after radiotherapy (Shaverdian et al. 2017; Grimaldi et al. 2014). Currently, the combination of pembrolizumab with or without stereotactic body radiation therapy (SBRT), and the combination of nivolumab and ipilimumab with or without SBRT, in advanced MCC, are under investigation (NCT03304639 and NCT03071406) (See Table 2).

What after immunotherapy?

Patients not responding to immunotherapy

For those patients who do not respond to immunotherapy, other approaches are being explored for MCC.

The intratumoural immune infiltration of pro- and anti-inflammatory molecules stimulating the immune system has been evaluated. Glucopyranosyl lipid-A stable emulsion (GLA-SE), a Toll-like receptor 4, was reported to be associated with a CR in one patient and a quite long recurrence free period in patients with stage IIIB MCC, together with a partial response (PR) in 2 out of 7 patients with stage IV MCC (Bhatia et al. 2016). Other cytokines such as IL-2 and TNF-α were also injected in the MCC lesion with a 30% of CR and 10% of PR, while 40% of patients showed stable disease (SD) (Bhatia et al. 2016). The use of TTI-621, a recombinant fusion protein targeting CD 47 (NCT02890368) is being tested in an early phase trial.

An interesting strategy for advanced MCC not responding to ICI is the use of a Natural Killer (NK) cells based therapy, which preliminarily provided a CR in one patient (NCT02465957). The infusion of T cells plus HLA upregulation was also evaluated without benefits when administered alone, while three CR were observed when added in combination with avelumab (Chapuis et al. 2014; Paulson et al. 2017). Other clinical trials on T cell regulation therapy are under investigation (NCT03241173 and NCT03071757).

Another strategy is based on the use of mutated viral genes, which active proliferative pathways in tumour cells allowing the virus to replicate and kill these cells (Andtbacka et al. 2015). Other agents with similar activity are being explored such as the intralesional talimogene laherparepvec, a recombinant herpes simplex type-1 virus-based agent (NCT02819843, NCT03458117 and NCT02978625).

Further treatment strategies include targeted therapies, inhibiting molecules required for tumour growth and progression. Some of them are based on tropomyosin receptor kinase A expression or AKT/PI3K pathway, mTOR complexes, tyrosine kinases, such as VEGF and PDGF receptors, or survivin protein (Shiver et al. 2015; Davids et al. 2009; Samlowski et al. 2010; Wehkamp et al. 2017; Iwasaki et al. 2015; Arora et al. 2012; Brunner et al. 2008). Recently, the use of cabozantinib in patients with advanced, progressive MCC after platinum-based chemotherapy did not demonstrate any benefit in terms of disease control (Rabinowits et al. 2018). However, a recent case series of five patients treated with pazopanib or cabozantinib showed a clinical benefit with a stabilization of disease (Tarabadkar et al. 2018). In addition, preliminary results from a clinical trial investigating pazopanib showed a PR rate of 19% and a disease control rate (OR + SD for > 12 weeks) of 56% (clinicaltrialsregister.eu; Eudra CT# 2011-003226-27) (Nathan et al. 2016). Currently, clinical trials on cabozantinib for MCC are ongoing (NCT02036476).

The overexpression of somatostatin receptor 2 has been reported in patients with MCC (Guitera-Rovel et al. 2001) representing a potential target for both imaging and treatment (Buder et al. 2014; Meier et al. 2004). However, results from studies evaluating the efficacy of somatostatin analogues in MCC are not available, yet (NCT 02351128) (See Table 2).

Other immune checkpoint inhibitors

The use of antibodies blocking the CTLA4 expression (ipilimumab) in T cells can be considered for those patients where PD-1 pathway blockade is ineffective, trying to improve the immune response. Clinical trials investigating ipilimumab efficacy in MCC are ongoing (NCT02488759, NCT02196961 and NCT03071406).

Further, atezolizumab, durvalumab, tremelimumab and daratumumab, human IgG1 anti-PD-L1 monoclonal antibodies, are currently under investigation for the treatment of advanced MCC (NCT02471846, NCT02488759 and NCT02643303).

Interesting results come from the evaluation of utomilumab, a human IgG2 agonist monoclonal antibody that showed a favourable response in 2 patients with advanced MCC who achieved a CR (one patient) and a PR (the other) with a DoR > 6 and 22 months, respectively (Segal et al. 2018).

Finally, combinations of immunotherapeutic options are being evaluating (NCT02584829, NCT03212404 and NCT03000257)(See Table 2).

Patients who develop resistance or escape to immunotherapy

Despite high antitumour activity of antibodies directed against PD-1 immune checkpoint, resistance to this therapy is frequent due to the activation of adaptive resistance, with upregulation of alternative immune checkpoints. A recent study demonstrated that the upregulation of T-cell immunoglobulin muscin-3 (TIM-3) was associated with anti-PD-1 resistance, and the inhibition of TIM-reverted the resistance to anti-PD1 antibodies in a mouse model of lung adenocarcinoma (Koyama et al. 2016).

On the other hand, a possible mechanism of escape from immunotherapy is the loss of classical and non-classical major histocompatibility complex molecules impairing adaptative and innate immune responses (Ritter et al. 2016), that can be reversed by interferon or epigenetic modulation (Terheyden and Becker 2017).

Radiotherapy and interferon have been demonstrated to release tumour antigens and reverse immune escape mechanisms such as the downregulation of major histocompatibility complex 1, in patients with MCC (Paulson et al. 2014).

Rechallenge after discontinuation

The efficacy of ICI for the advanced MCC setting and the limited therapeutic options after progression raise the question if patients previously treated with these drugs, who experience a progression, can be retreated with the same or different agents, after discontinuation (Saleh et al. 2018).

To date, no available data exist for patients with MCC. However, some information on the rechallenge with targeted therapies is available for patients with other endocrine tumours (Felicetti et al. 2017). In advanced melanoma, a clinical benefit has been shown after discontinuation of nivolumab and following rechallenge (Long et al. 2017), with sustained PR (Lipson et al. 2013). However, in a recent case series, the rechallenge with anti-PD-1 was associated with a progressive disease (Martini et al. 2017), while Blasig et al. reported a good response in a series of eight patients, with a SD in three patients and a PR in one patient (Blasig et al. 2017).

Anyway, the development of resistance to any anti-PD1/PD-L1 may lead to resistance to other agents with the same mechanism of action.

Chemotherapy

Due to the effective role of immunotherapy and the reduced risk of side effects, in the next years chemotherapy will remain an option only for those patients with metastatic MCC who are not candidates for immunotherapy or show a progression after immunotherapy. Furthermore, chemotherapy may be reserved for metastatic MCC patients with high tumour burden, acute tumour related symptoms and a good performance status, requiring a fast response.

The impact of chemotherapy on the expression of PD-L1 and PD-1 in cell lines has already been investigated, with different results depending on the experimental model and the chemotherapeutic agent investigated (Chacon et al. 2016; Ghebeh et al. 2010; Peng et al. 2015; Zhang et al. 2008). A recent study evaluated the expression of PD-L1/PD1 in tumour and immune cells of lung cancer after platinum-based chemotherapy, showing that neoadjuvant chemotherapy decreases PD-L1 expression of tumour cells, but not of immune cells, requiring a re-evaluation of PD-L1 expression before immunotherapy (Rojkó et al. 2018). In addition, a significant downregulation of PD-L1 expression with cisplatin–gemcitabine regimen and a tendency to upregulation with carboplatin–paclitaxel regimen, was reported.

In line with Rojkò et al. results, Sheng et al. and Zhang et al. have shown that neoadjuvant chemotherapy induces resistance to immunotherapy, decreasing PD-L1 expression in patients with NSCLC (Sheng et al. 2016; Zhang et al. 2016).

Interesting results from a study on patients with Hodgkin lymphoma treated with chemotherapy alone after PD with ICI showed a 46% of CR, supporting the hypothesis that immunotherapy may sometimes re-sensitize patient response to chemotherapy (Rossi et al. 2018), even though further studies are needed to confirm these results.

Currently, however, no data are available on the use of chemotherapy after immunotherapy in MCC, even though the combination of these agents is being evaluating (NCT 03167164) (See Table 2).

Immunotherapy in patients excluded from clinical trials

The safety and efficacy of anti-PD-1/PD-L1 therapy has been investigated in several clinical trials. However, some conditions often associated to a higher incidence of MCC, such as organ transplantation, HIV infection, active autoimmune diseases, and hematologic or solid malignancies, are commonly considered as exclusion criteria for clinical trials patient’s enrollment. Therefore, little experience exists in this setting, except for clinical case reports and small cohort studies.

Autoimmune toxicity represents the most frequent side effect of ICIs, as a consequence of T cells activation against host tissue. For this reason, subjects with autoimmune diseases were excluded from clinical trials, as the further immune stimulation produced by ICIs could lead to an exacerbation of autoimmune disorders and related symptoms (Khan et al. 2016). An interesting study on 52 patients with melanoma and preexisting autoimmune disease showed that treatment with anti-PD 1 triggered autoimmune flare in 30% of patients, resulting in 4% treatment discontinuations, and caused immune related adverse events (irAEs) in another 29% of them, leading to therapy interruption with a rate of 8%. In this cohort, treatment with anti-PD 1 was effective only in one-third of patients (Johnson et al. 2016). In another retrospective study on 41 patients with advanced melanoma and preexisting autoimmune disorders or ipilimumab-induced autoimmune toxicity, 8 patients experienced a flare of autoimmune disease, requiring immunosuppressive therapy (Johnson et al. 2016).

Traditionally, recipients of solid or hematologic stem cells transplants are treated with immunosuppressive drugs to maintain allograft tolerance and to prevent graft versus host disease (GVHD). However, chronic immunosuppression represents a risk factor for the development of a wide spectrum of malignancies, such as MCC (Engels et al. 2011). Despite the increasing need for effective cancer treatments, ICIs are currently contraindicated in organ transplanted patients, because of the higher risk of organ rejection. Accordingly, preclinical data suggested that PD1/PD-L1 could be mainly involved in organ tolerance and that blockage of the PD-1 pathway may lead to an increased organ transplant rejection rate (Terawaki et al. 2007; Herz et al. 2016). Conversely, ICIs have been used in patients following hematopoietic stem cells transplantion without experiencing a higher rate of side effects (Angenendt et al. 2016; Villasboas et al. 2016; Yared et al. 2016). Immunotherapy was also used as a salvage therapy in different transplanted patients, but organ rejection has been frequent in this setting (Barnett et al. 2017; De Toni and Gerbes 2017; Kittai et al. 2017). Fourteen patients who received liver transplant were treated by different ICIs and liver graft rejection was reported in four of them, in 3 cases with lethal outcome. A review of 12 case reports of transplanted patients (9 kidney transplants, 2 liver transplants and 1 heart transplant) treated by anti CTLA 4 or anti PD-1 therapy, mostly for melanoma (7 patients), confirmed a high incidence of graft rejection. Indeed 4 patients, all after kidney transplant, developed a definitive organ rejection. However, 8 patients experienced a PR or SD (duration of response between 4 months and 2,5 years). Kidney transplant was associated to a higher incidence of graft rejection, while a longer time elapsed since transplantation was associated with a higher tolerance (Kittai et al. 2017).

HIV positive patients represent another category of patients for whom ICIs are considered contraindicated, due to the risk of exacerbating the infection or to develop irAEs. In a case series including 10 patients with HIV infection, on antiretroviral therapy, the use of ICIs (3 patients treated with pembrolizumab, 1 with nivolumab, 3 with ipilimumab, and 3 with ipilimumab and nivolumab) for advanced melanoma (9 patients) and MCC (1 patient) did not significantly affect the viral load. Treated patients experienced one CR and one PR among patients treated for melanoma, and a CR in the patient treated for MCC. Toxicities were represented by irAEs in 50% of patients, which were severe in 2 cases (colitis and myositis) leading to treatment discontinuation (Heppt et al. 2017). Another case report showed a CR in a 88-year-old, HIV-infected patients, treated with avelumab for MCC resistant to chemotherapy and radiotherapy. At the time of the publication, the patient received ten administrations of avelumab and experienced hypothyroidism due to an autoimmune thyroiditis (Al Homsi et al. 2018). Even though it is a clinical case-based evidence, ICIs seem to display the same efficacy in HIV positive patients.

Patients with HBV and HCV infections were also excluded from clinical trials. A retrospective study showed that pembrolizumab (2 patients) and nivolumab (5 patients) were safely used also in these patients for the treatment of melanoma or of NSCLC. Only one patient experienced a grade 2 liver toxicity (according to CTCAEs), treated with ledipasvir and sofosbuvir. Two patients experienced a PR and 2 patients a durable SD, still ongoing at the time of publication. However, patients with active hepatitis require a close hepatologic monitoring during treatment (Kothapalli and Khattak 2018).

ICIs, as monoclonal antibodies, are metabolized by circulating fagocitic cells and do not undergo hepatic or renal clearance, thus liver or renal failure do not represent treatment contraindications (Mould and Sweeney 2007). However, patients with severe renal, liver or cardiac dysfunction were excluded from clinical trials. Kanz et al. treated with anti PD-1/PD-L1 27 patients with renal (creatinine ≥ 2 mg/dl or creatinine clearance < 30 ml/min), hepatic (AST, ALT or bilirubine ≥ 3 × upper limit) or cardiac (ejection fraction < 45%) dysfunction, showing a similar incidence and kind of adverse events and a durable response in several patients (Kanz et al. 2016). Similar results were reported by other studies (El-Khoueiry et al. 2017; Rosenberg et al. 2016).

Finally, a recent phase II trial assessed the efficacy of pembrolizumab in patients with untreated or progressing brain metastases from melanoma or NSCLC (18 patients). Data showed the efficacy of pembrolizumab on brain metastases in 33% of patients, with a CR in 4 patients and a PR in 2 patients. Treatment was well tolerated with grade ≤ 2 neurologic toxicity (Goldberg et al. 2016).

Summarizing, ICIs demonstrated a good safety and efficacy profile also in patients classically excluded from clinical trials, suggesting that these treatments could be safely used also in these patients, that are representative of the “real world”. Organ transplanted patients represent an exception, due to the high risk of organ rejection. However, larger prospective studies could better investigate this issue.

How long should treatment with immune checkpoint inhibitors be prolonged after complete response?

According to the American Society of Clinical Oncology (ASCO) and the European Society of Medical Oncology (ESMO), as well as to the summaries of product characteristics, avelumab and pembrolizumab should be administrated until disease progression or unacceptable toxicity (Brahmer et al. 2018; Haanen et al. 2018; BAVENCIO RCP 2018; KEYTRUDA RCP 2018), but in the real-life clinical practice the optimal duration of ICIs therapy is still unknown. In particular, few data are available regarding the ideal duration of therapy with avelumab in MCC in cases of complete response (CR). In a recent multicentre, single-group, open-label, phase II trial, complete responders were treated for a minimum of 6 months till a maximum of 12 months, after CR detection, based on investigator’s discretion (Kaufman et al. 2016).

However, some evidence is already available about two anti-PD-1 ICIs (nivolumab and pembrolizumab) especially for patients with advanced melanoma. In a randomized, controlled, phase III study (Robert et al. 2015) the administration of pembrolizumab or ipilimumab was arbitrarily stopped after 2 years of treatment, without any specific evidence supporting that. The KEYNOTE-001 study report (Robert et al. 2016), published in 2016, showed a CR in 95 patients with advanced melanoma treated with pembrolizumab. Among these, 61 patients discontinued therapy because of CR and two of them relapsed after discontinuation. In 2017, a retrospective review of CR to PD-1 based therapy in patients with metastatic melanoma was published (Ladwa and Atkinson 2017). The authors investigated two groups of patients treated with pembrolizumab (the Pembrolizumab Named Patient Program [PEM NPP, N = 20] and reimbursed Pembrolizumab [r PEM, N = 3]), and one group treated with nivolumab monotherapy (the NIVO, N = 6). Among these patients that discontinued the treatment because of CR, 3 relapsed at a median follow-up of 8 months. The main limitations of this study are the small number of patients and the short follow-up after discontinuation (10 months in PEM NPP, 4.5 months in PEM, and 9 months in NIVO). However, the interesting aspect is that all the patients who had relapsed were successfully retreated with immunotherapy (Ladwa and Atkinson 2017). A recent report from the KEYNOTE-006 study, which was presented at the 2018 ASCO Congress (Long et al. 2018), evaluated the 4-year OS, PFS, and clinical outcomes in patients with ipilimumab-naive advanced melanoma treated with ipilimumab or pembrolizumab for 2 years. Pembrolizumab performed better than ipilimumab in terms of OS, PFS and duration of response, and after 4 years of follow-up it continues to provide durable antitumour activity (Long et al. 2018). Moreover, the rechallenge with ICIs upon disease progression can provide additional antitumour activity with acceptable safety (Ladwa and Atkinson 2017; Long et al. 2018).

Besides costs, another issue to take into account for ICIs is the treatment-related toxicity, especially because MCC mainly affects the elderly. Avelumab was found to be well tolerated (Kelly et al. 2018) especially if compared to chemotherapy, however, no data are available regarding the safety of long-term therapies.

In conclusion, it could be speculated that prolonging therapy with ICIs beyond 24 months in patients with CR may be not required, considering costs, the potential toxicity, and the possibility to retreat successfully the patients.

What follow-up should be performed during treatment and after complete response?

About the timing of follow-up, the NCCN Clinical Practice guidelines recommend physical exam with complete skin and lymph node evaluation every 3–6 months for the first 3 years after diagnosis, and every 6–12 months thereafter, although individuals at high risk of recurrence (e.g. immunosuppressed patients) should deserve more frequent follow-up (Bichakjian et al. 2018).

The follow-up of these patients should also include the screening of irAEs of ICIs, which most commonly affect the skin, the gastrointestinal tract, the liver, and the endocrine system, although other organs may also be affected (Postow 2015a, b; Boutros et al. 2016). Particularly, endocrine irAEs include hypothyroidism/hyperthyroidism, hypophysitis and autoimmune insulin-dependent diabetes mellitus, with a toxicity which is potentially irreversible (Orlov et al. 2015).

For ipilimumab, nivolumab and pembrolizumab studies in melanoma patients, the median time to onset of moderate to severe endocrine irAEs was 2–5 months after the initiation of therapy (Weber et al. 2013; Faje et al. 2014; OPDIVO RCP 2018; KEYTRUDA RCP 2018), but toxicity can appear even beyond the final dose of treatment, which makes it necessary to continue follow-up for several months after cessation of treatment.

To identify and quantify regional and distant metastases after CR, the NCCN guidelines for MCC recommend contrast-enhanced brain MRI and contrast-enhanced neck/chest/abdomen/pelvis CT or whole body 18FDG PET/CT (Bichakjian et al. 2018). Some studies indicated that both whole body 18FDG PET/CT and 68Ga-somatostatin analogue PET/CT can be useful for monitoring the response of metastases of MCC to active immunotherapy (Eshghi et al. 2018; Taralli et al. 2018).

Conclusions and future perspectives

The development of ICIs has recently revolutionised the clinical management and the prognosis of MCC. Paradoxically, however, many of the patients at higher risk of developing this rare, albeit highly aggressive neuroendocrine tumour seem to be excluded from the possibility to be treated with ICIs.

Several questions on the treatment of patients with MCC are still unresolved. In this manuscript, we tried to deal with some of the most relevant unanswered issues on this topic. However, many other aspects are already facing out on the horizon. What is the role of the disease mutational burden on the response to immunotherapy? Can immunomodulatory agents (e.g. somatostatin analogues) or the combination with radiotherapy help ICIs modulating lymphocyte response of the disease? Should the dose of ICIs be modulated according to the patient characteristics or does a single dose fits all? Will precision medicine help us finding the least toxic and/or most effective drug/dose to tailor the individual patient with MCC? Will the findings of ICIs on MCC pave the way for similar achievements in some other neuroendocrine tumours?

Indeed, preliminary data from the multicohort phase 1b KEYNOTE-28 study (NCT02054806) already showed promising results with pembrolizumab in heavily pretreated patients with PD-L1 positive carcinoid tumours and with well- or moderately differentiated pancreatic neuroendocrine tumours (Menhert et al. 2017).

The therapeutic landscape of neuroendocrine tumours, and in particular of MCC, is rapidly evolving: many open issues will probably be resolved, and many other questions are likely to arise in the next few years. The results of ongoing prospective clinical trials and of several other studies on these issues are eagerly awaited. In the meanwhile, it is reasonable to expect that some other answers to these open issues will not be late.

Acknowledgements

This review is part of the ‘NIKE’ project (Neuroendocrine tumours Innovation Knowledge and Education) led by Prof Annamaria Colao, which aims at increasing the knowledge on NETs. We would like to acknowledge all the Collaborators of this project: Manuela Albertelli, Barbara Altieri, Elena Ambrosetti, Antonio Bianchi, Lena Bottiglieri, Severo Campione, Silvia Carra, Carla Di Dato, Sergio Di Molfetta, Antonella Di Sarno, Giuseppe Fanciulli, Diego Ferone, Francesco Ferraù, Elisa Giannetta, Federica Grillo, Erika Maria Grossrubatscher, Elia Guadagno, Andrea Isidori, Fabio Lo Calzo, Pasqualino Malandrino, Chiara Martini, Erika Messina, Roberta Modica, Giovanna Muscogiuri, Genoveffa Pizza, Paola Razzore, Laura Rizza, Rosaria Maddalena Ruggeri, Concetta Sciammarella, Giovanni Vitale, and Maria Chiara Zatelli.

Author contributions

All authors have contributed equally to the conception and design of the review. MG, FDC, VG, and MR analyzed data and drafted the article. AF and AC revised the manuscript critically. All authors approved the final article.

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Compliance with ethical standards

Conflict of interest

The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.

Ethical approval

This article does not contain any studies with human participants or animals performed by any of the authors.

Footnotes

The members of the NIKE Group are given in the Acknowledgement section.

Contributor Information

Marco Gallo, Phone: +39-011-6336611, Email: mgallo4@cittadellasalute.to.it.

NIKE Group:

Manuela Albertelli, Barbara Altieri, Elena Ambrosetti, Antonio Bianchi, Lena Bottiglieri, Severo Campione, Silvia Carra, Carla Di Dato, Sergio Di Molfetta, Antonella Di Sarno, Giuseppe Fanciulli, Diego Ferone, Francesco Ferraù, Elisa Giannetta, Federica Grillo, Erika Maria Grossrubatscher, Elia Guadagno, Andrea Isidori, Fabio Lo Calzo, Pasqualino Malandrino, Chiara Martini, Erika Messina, Roberta Modica, Giovanna Muscogiuri, Genoveffa Pizza, Paola Razzore, Laura Rizza, Rosaria Maddalena Ruggeri, Concetta Sciammarella, Giovanni Vitale, and Maria Chiara Zatelli

References

  1. Al Homsi MU, Mostafa M, Fahim K (2018) Favorable response to treatment with avelumabin an HIV-positive patient with advanced merkel cell carcinoma previously refractory to chemotherapy. Case Rep Oncol 11(2):467–475. 10.1159/000490636 [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Albores-Saavedra J, Batich K, Chable-Montero F, Sagy N, Schwartz AM, Henson DE (2010) Merkel cell carcinoma demographics, morphology, and survival based on 3870 cases: a population based study. J Cutan Pathol 37:20–27. 10.1111/j.1600-0560.2009.01370.x [DOI] [PubMed] [Google Scholar]
  3. Amaral T, Leiter U, Garbe C (2017) Merkel cell carcinoma: epidemiology, pathogenesis, diagnosis and therapy. Rev Endocr Metab Disord 18:517–532. 10.1007/s11154-017-9433-0 [DOI] [PubMed] [Google Scholar]
  4. Andtbacka RH, Kaufman HL, Collichio F, Amatruda T, Senzer N, Chesney J et al (2015) Talimogene Laherparepvec improves durable response rate in patients with advanced melanoma. J Clin Oncol 33(25):2780–2788. 10.1200/JCO.2014.58.3377 [DOI] [PubMed] [Google Scholar]
  5. Angenendt L, Schliemann C, Lutz M, Rebber E, Schulze AB, Weckesser M et al (2016) Nivolumab in a patient with refractory Hodgkin’s lymphoma after allogeneic stem cell transplantation. Bone Marrow Transpl 51(3):443–445. 10.1038/bmt.2015.266 [DOI] [PubMed] [Google Scholar]
  6. Arora R, Shuda M, Guastafierro A, Feng H, Toptan T, Tolstov Y et al (2012) Survivin is a therapeutic target in Merkel cell carcinoma. Sci Transl Med 4(133):133ra56. 10.1126/scitranslmed.3003713 [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Baker M, Cordes L, Brownell I (2018) Avelumab: a new standard for treating metastatic Merkel cell carcinoma. Expert Rev Anticancer Ther 18:319–326. 10.1080/14737140.2018.1445528 [DOI] [PubMed] [Google Scholar]
  8. Barnett R, Barta VS, Jhaveri KD (2017) Preserved renal-allograft function and the PD-1 pathway inhibitor nivolumab. N Engl J Med 376(2):191–192. 10.1056/NEJMc1614298 [DOI] [PubMed] [Google Scholar]
  9. BAVENCIO (avelumab) (2018) Summary of product characteristics. http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Product_Information/human/004338/WC500236647.pdf. Accessed 18 Oct 2018
  10. Becker JC, Lorenz E, Ugurel S, Eigentler TK, Kiecker F, Pföhler C et al (2017) Evaluation of real-world treatment outcomes in patients with distant metastatic Merkel cell carcinoma following second-line chemotherapy in Europe. Oncotarget 8:79731–79741. 10.18632/oncotarget.19218 [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Becker JC, Stang A, Hausen AZ, Fischer N, DeCaprio JA, Tothill RW et al (2018) Epidemiology, biology and therapy of Merkel cell carcinoma: conclusions from the EU project IMMOMEC. Cancer Immunol Immunother 67:341–351. 10.1007/s00262-017-2099-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Bhatia S, Miller N, Lu H, Ibrani D, Shinohara M, Byrd DR et al (2016) Pilot trial of intratumoral (IT) G100, a toll-like receptor-4 (TLR4) agonist, in patients (pts) with Merkel cell carcinoma (MCC): Final clinical results and immunologic effects on the tumor microenvironment (TME). 2016 ASCO Annual Meeting. J Clin Oncol 34(15 suppl):3021 [Google Scholar]
  13. Bichakjian CK, Nghiem P, Johnson T et al (2017) Merkel cell carcinoma. In: Amin MB, Edge SB, Greene FL et al (eds) AJCC cancer staging manual, 8th edn. Springer, New York, pp 549–562 [Google Scholar]
  14. Bichakjian CK, Olencki T, Aasi SZ, Alam M, Andersen JS, Blitzblau R et al (2018) Merkel cell carcinoma, Version 1.2018, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw 16:742–774. 10.6004/jnccn.2018.0055 [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Blasig H, Bender C, Hassel JC, Eigentler TK, Sachse MM, Hiernickel J et al (2017) Reinduction of PD1-inhibitor therapy: first experience in eight patients with metastatic melanoma. Melanoma Res 27(4):321–325. 10.1097/CMR.0000000000000341 [DOI] [PubMed] [Google Scholar]
  16. Bommareddy PK, Kaufman HL (2017) Avelumab and other recent advances in Merkel cell carcinoma. Future Oncol 13:2771–2783. 10.2217/fon-2017-0305 [DOI] [PubMed] [Google Scholar]
  17. Boutros C, Tarhini A, Routier E, Lambotte O, Ladurie FL, Carbonnel F et al (2016) Safety profiles of anti-CTLA-4 and anti-PD-1 antibodies alone and in combination. Nat Rev Clin Oncol 13:473–486. 10.1038/nrclinonc.2016.58 [DOI] [PubMed] [Google Scholar]
  18. Brahmer JR, Lacchetti C, Schneider BJ, Atkins MB, Brassil KJ, Caterino JM et al (2018) National comprehensive cancer network. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol 36(17):1714–1768. 10.1200/JCO.2017.77.6385 [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Brunner M, Thurnher D, Pammer J, Geleff S, Heiduschka G, Reinisch CM et al (2008) Expression of VEGF-A/C, VEGF-R2, PDGF-alpha/beta, c-kit, EGFR, Her-2/Neu, Mcl-1 and Bmi-1 in Merkel cell carcinoma. Mod Pathol 21(7):876–884. 10.1038/modpathol.2008.63 [DOI] [PubMed] [Google Scholar]
  20. Buder K, Lapa C, Kreissl MC, Schirbel A, Herrmann K, Schnack A et al (2014) Somatostatin receptor expression in Merkel cell carcinoma as target for molecular imaging. BMC Cancer 14:268. 10.1186/1471-2407-14-268 [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Cassler NM, Merrill D, Bichakjian CK, Brownell I (2016) Merkel cell carcinoma therapeutic update. Curr Treat Options Oncol 17:36. 10.1007/s11864-016-0409-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Chacon JA, Schutsky K, Powell DJ (2016) The impact of chemotherapy, radiation and epigenetic modifiers in cancer cell expression of immune inhibitory and stimulatory molecules and anti-tumor efficacy. Vaccines 4(4):43. 10.3390/vaccines4040043 [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Chapuis AG, Afanasiev OK, Iyer JG, Paulson KG, Parvathaneni U, Hwang JH et al (2014) Regression of metastatic Merkel cell carcinoma following transfer of polyomavirus-specific T cells and therapies capable of re-inducing HLA class-I. Cancer Immunol Res 2:27–36. 10.1158/2326-6066.CIR-13-0087 [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Clarke CA, Robbins HA, Tatalovich Z, Lynch CF, Pawlish KS, Finch JL (2015) Risk of Merkel cell carcinoma after solid organ transplantation. J Natl Cancer Inst. 10.1093/jnci/dju382 [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. D’Angelo SP, Russell J, Lebbé C, Chmielowski B, Gambichler T, Grob JJ et al (2018) Efficacy and Safety of first-line avelumab treatment in patients with stage IV metastatic Merkel cell carcinoma: a preplanned interim analysis of a clinical trial. JAMA Oncol. 10.1001/jamaoncol.2018.0077 [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Davids MS, Charlton A, Ng SS, Chong ML, Laubscher K, Dar M et al (2009) Response to a novel multitargeted tyrosine kinase inhibitor pazopanib in metastatic Merkel cell carcinoma. J Clin Oncol 27(26):e97–e100. 10.1200/JCO.2009.21.8149 [DOI] [PubMed] [Google Scholar]
  27. De Toni EN, Gerbes AL (2017) Tapering of immunosuppression and sustained treatment with nivolumab in a liver transplant recipient. Gastroenterology 152(6):1631–1633. 10.1053/j.gastro.2017.01.063 [DOI] [PubMed] [Google Scholar]
  28. Demaria S, Ng B, Devitt ML, Babb JS, Kawashima N, Liebes L et al (2004) Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Int J Radiat Oncol Biol Phys 58:862–870. 10.1016/j.ijrobp.2003.09.012 [DOI] [PubMed] [Google Scholar]
  29. Deneve JL, Messina JL, Marzban SS, Gonzalez RJ, Walls BM, Fisher KJ et al (2012) Merkel cell carcinoma of unknown primary origin. Ann Surg Oncol 19:2360–2366. 10.1245/s10434-011-2213-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Deng L, Liang H, Burnette B, Beckett M, Darga T, Weichselbaum RR et al (2014) Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J Clin Invest 124:687–695. 10.1172/JCI67313 [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Eggermont AMM, Chiarion-Sileni V, Grob JJ, Dummer R, Wolchok JD, Schmidt H et al (2015) Adjuvant ipilimumab versus placebo after complete resection of high-risk stage III melanoma (EORTC 18071): a randomised, double-blind, phase 3 trial. Lancet Oncol 16:522–530. 10.1016/S1470-2045(15)70122-1 [DOI] [PubMed] [Google Scholar]
  32. Eggermont AMM, Blank CU, Mandala M, Long GV, Atkinson V, Dalle S et al (2018) Adjuvant pembrolizumab versus placebo in resected stage III melanoma. N Engl J Med 378:1789–1801. 10.1056/NEJMoa1802357 [DOI] [PubMed] [Google Scholar]
  33. El-Khoueiry AB, Sangro B, Yau T, Crocenzi TS, Kudo M, Hsu C et al (2017) Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet 389(10088):2492–2502. 10.1016/S0140-6736(17)31046-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Engels EA, Frisch M, Goedert JJ, Biggar RJ, Miller RW (2002) Merkel cell carcinoma and HIV infection. Lancet 359:497–498. 10.1016/S0140-6736(02)07668-7 [DOI] [PubMed] [Google Scholar]
  35. Engels EA, Pfeiffer RM, Fraumeni JF Jr, Kasiske BL, Israni AK, Snyder JJ et al (2011) Spectrum of cancer risk among US solid organ transplant recipients. JAMA 306(17):1891–1901. 10.1001/jama.2011.1592 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Eshghi N, Lundeen TF, MacKinnon L, Avery R, Kuo PH (2018) 18F-FDG PET/CT for monitoring response of Merkel cell carcinoma to the novel programmed cell death ligand 1 inhibitor avelumab. Clin Nucl Med. 10.1097/RLU.0000000000002051 [DOI] [PubMed] [Google Scholar]
  37. Faje AT, Sullivan R, Lawrence D, Tritos NA, Fadden R, Klibanski A et al (2014) Ipilimumab-induced hypophysitis: a detailed longitudinal analysis in a large cohort of patients with metastatic melanoma. J Clin Endocrinol Metab 99:4078–4085. 10.1210/jc.2014-2306 [DOI] [PubMed] [Google Scholar]
  38. Felicetti F, Nervo A, Piovesan A, Berardelli R, Marchisio F, Gallo M et al (2017) Tyrosine kinase inhibitors rechallenge in solid tumors: a review of literature and a case description with lenvatinib in thyroid cancer. Expert Rev Anticancer Ther 17(12):1093–1098. 10.1080/14737140.2017.1390432 [DOI] [PubMed] [Google Scholar]
  39. Forde PM, Chaft JE, Smith KN, Anagnostou V, Cottrell TR, Hellmann MD et al (2018) Neoadjuvant PD-1 blockade in resectable lung cancer. N Engl J Med 378:1976–1986. 10.1056/NEJMoa1716078 [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Ghebeh H, Lehe C, Barhoush E, Al-Romaih K, Tulbah A, Al-Alwan M et al (2010) Doxorubicin downregulates cell surface B7-H1 expression and upregulates its nuclear expression in breast cancer cells: role of B7-H1 as an anti-apoptotic molecule. Breast Cancer Res BCR 12(4):R48. 10.1186/bcr2605 https [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Gibney GT, Kudchadkar RR, DeConti RC, Thebeau MS, Czupryn MP, Tetteh L et al (2015) Safety, correlative markers, and clinical results of adjuvant nivolumab in combination with vaccine in resected high-risk metastatic melanoma. Clin Cancer Res 21:712–720. 10.1158/1078-0432.CCR-14-2468 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Goh G, Walradt T, Markarov V, Blom A, Riaz N, Doumani R et al (2016) Mutational landscape of MCPyV-positive and MCPyV-negative Merkel cell carcinomas with implications for immunotherapy. Oncotarget 7:3403–3415. 10.18632/oncotarget.6494 [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Goldberg SB, Gettinger SN, Mahajan A, Chiang AC, Herbst RS, Sznol M et al (2016) Pembrolizumab for patients with melanoma or non-small-cell lung cancer and untreated brain metastases: early analysis of a non-randomised, open-label, phase 2 trial. Lancet Oncol 17(7):976–983. 10.1016/S1470-2045(16)30053-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Grimaldi AM, Simeone E, Giannarelli D, Muto P, Falivene S, Borzillo V et al (2014) Abscopal effects of radiotherapy on advanced melanoma patients who progressed after ipilimumab immunotherapy. Oncoimmunology 3:e28780. 10.4161/onci.28780 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Guitera-Rovel P, Lumbroso J, Gautier-Gougis MS, Spatz A, Mercier S, Margulis S et al (2001) Indium-111octreotide scintigraphy of Merkel cell carcinomas and their metastases. Ann Oncol 12:807–811 [DOI] [PubMed] [Google Scholar]
  46. Haanen JBAG, Carbonnel F, Robert C, Kerr KM, Peters S, Larkin J et al (2018) ESMO Guidelines Committee. Management of toxicities from immunotherapy: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 10.1093/annonc/mdy162 [DOI] [PubMed] [Google Scholar]
  47. Hasan S, Liu L, Triplet J, Li Z, Mansur D (2013) The role of postoperative radiation and chemoradiation in Merkel cell carcinoma: a systematic review of the literature. Front Oncol 3:276. 10.3389/fonc.2013.00276 [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Hauschild A, Schadendorf D (2016) Checkpoint inhibitors: a new standard of care for advanced Merkel cell carcinoma? Lancet Oncol 17:1337–1339. 10.1016/S1470-2045(16)30441-7 [DOI] [PubMed] [Google Scholar]
  49. Heppt MV, Schlaak M, Eigentler TK, Kähler KC, Kiecker F, Loquai C et al (2017) Checkpoint blockade for metastatic melanoma and Merkel cell carcinoma in HIV-positive patients. Ann Oncol 28(12):3104–3106. 10.1093/annonc/mdx538 [DOI] [PubMed] [Google Scholar]
  50. Herz S, Höfer T, Papapanagiotou M, Leyh JC, Meyenburg S, Schadendorf D et al (2016) Checkpoint inhibitors in chronic kidney failure and an organ transplant recipient. Eur J Cancer 67:66–72. 10.1016/j.ejca.2016.07.026 [DOI] [PubMed] [Google Scholar]
  51. Hodgson NC (2005) Merkel cell carcinoma: changing incidence trends. J Surg Oncol 89:1–4. 10.1002/jso.20167 [DOI] [PubMed] [Google Scholar]
  52. Iwasaki T, Matsushita M, Nonaka D, Kuwamoto S, Kato M, Murakami I et al (2015) Comparison of Akt/mTOR/4E-BP1 pathway signal activation and mutations of PIK3CA in Merkel cell polyomavirus-positive and Merkel cell polyomavirus-negative carcinomas. Hum Pathol 46(2):210–216. 10.1016/j.humpath.2014.07.025 [DOI] [PubMed] [Google Scholar]
  53. Iyer JG, Blom A, Doumani R, Lewis C, Tarabadkar ES, Anderson A et al (2016) Response rates and durability of chemotherapy among 62 patients with metastatic Merkel cell carcinoma. Cancer Med 5:2294–2301. 10.1002/cam4.815 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Johnson DB, Sullivan RJ, Ott PA, Carlino MS, Khushalani NI, Ye F et al (2016) Ipilimumab therapy in patients with advanced melanoma and preexisting autoimmune disorders. JAMA Oncol 2(2):234–240. 10.1001/jamaoncol.2015.4368 [DOI] [PubMed] [Google Scholar]
  55. Kanz BA, Pollack MH, Johnpulle R, Puzanov I, Horn L, Morgans A et al (2016) Safety and efficacy of anti-PD-1 in patients with baseline cardiac, renal, or hepatic dysfunction. J Immunother Cancer 4:60. 10.1186/s40425-016-0166-5 (eCollection 2016) [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Kaufman HL, Russell J, Hamid O, Bhatia S, Terheyden P, D’Angelo SP et al (2016) Avelumab in patients with chemotherapy-refractory metastatic Merkel cell carcinoma: a multicentre, single-group, open-label, phase 2 trial. Lancet Oncol 17:1374–1385. 10.1016/S1470-2045(16)30364-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Kaufman HL, Russell JS, Hamid O, Bhatia S, Terheyden P, D’Angelo SP et al (2018) 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. J Immunother Cancer 6:7. 10.1186/s40425-017-0310-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Kelly K, Infante JR, Taylor MH, Patel MR, Wong DJ, Iannotti N et al (2018) Safety profile of avelumab in patients with advanced solid tumors: A pooled analysis of data from the phase 1 JAVELIN solid tumor and phase 2 JAVELIN Merkel 200 clinical trials. Cancer. 10.1002/cncr.31293 [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. KEYTRUDA (pembrolizumab) (2018) Summary of product characteristics. https://www.ema.europa.eu/documents/product-information/keytruda-epar-product-information_en.pdf. Accessed 18 Oct 2018
  60. Khan SA, Pruitt SL, Xuan L, Gerber DE (2016) Prevalence of autoimmune disease among patients with lung cancer: implications for immunotherapy treatment options. JAMA Oncol 2(11):1507–1508. 10.1001/jamaoncol.2016.2238 [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Kittai AS, Oldham H, Cetnar J, Taylor M (2017) Immune checkpoint inhibitors in organ transplant patients. J Immunother 40(7):277–281. 10.1097/CJI.0000000000000180 [DOI] [PubMed] [Google Scholar]
  62. Kothapalli A, Khattak MA (2018) Safety and efficacy of anti-PD-1 therapy for metastatic melanoma and non-small-cell lung cancer in patients with viral hepatitis: a case series. Melanoma Res 28(2):155–158. 10.1097/CMR.0000000000000434 [DOI] [PubMed] [Google Scholar]
  63. Koyama S, Akbay EA, Li YY, Herter-Sprie GS, Buczkowski KA, Richards WG et al (2016) Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat Commun 7:10501. 10.1038/ncomms10501 [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Ladwa R, Atkinson V (2017) The cessation of anti-PD-1 antibodies of complete responders in metastatic melanoma. Melanoma Res 27(2):168–170. 10.1097/CMR.0000000000000336 [DOI] [PubMed] [Google Scholar]
  65. Lipson EJ, Sharfman WH, Drake CG, Wollner I, Taube JM, Anders RA et al (2013) Durable cancer regression off-treatment and effective reinduction therapy with an anti-PD-1 antibody. Clin Cancer Res 19(2):462–468. 10.1158/1078-0432.CCR-12-2625 [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Liu J, Blake SJ, Yong MC, Harjunpää H, Ngiow SF, Takeda K et al (2016) Improved efficacy of neoadjuvant compared to adjuvant immunotherapy to eradicate metastatic disease. Cancer Discov 6:1382–1399. 10.1158/2159-8290.CD-16-0577 [DOI] [PubMed] [Google Scholar]
  67. Long GV, Weber JS, Larkin J, Atkinson V, Grob JJ, Schadendorf D et al (2017) Nivolumab for patients with advanced melanoma treated beyond progression: analysis of 2 phase 3 clinical trials. JAMA Oncol 3(11):1511–1519. 10.1001/jamaoncol.2017.1588 [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Long GV, Schachter J, Ribas A, Arance A, Grob J-J, Mortier L et al (2018) 4-year survival and outcomes after cessation of pembrolizumab after 2 years in patients with ipilimumab-naive advanced melanoma in keynote-006. 2018 ASCO annual meeting. J Clin Oncol 36(15 suppl):9503 [Google Scholar]
  69. Martini DJ, Lalani AA, Bossé D, Steinharter JA, Harshman LC, Hodi FS et al (2017) Response to single agent PD-1 inhibitor after progression on previous PD-1/PD-L1 inhibitors: a case series. J Immunother Cancer 5(1):66. 10.1186/s40425-017-0273-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Meier G, Waldherr C, Herrmann R, Maecke H, Mueller-Brand J, Pless M (2004) Successful targeted radiotherapy with 90Y-DOTATOC in a patient with Merkel cell carcinoma. A case report. Oncology 66(2):160–163 [DOI] [PubMed] [Google Scholar]
  71. Menhert JM, Rugo HS, O’Neil BH, Santoro A, Schellens JHM, Cohen RB et al (2017) Pembrolizumab for patients with PD-L1–positive advanced carcinoid or pancreatic neuroendocrine tumors: Results from the KEYNOTE-028 study. Ann Oncol. 10.1093/annonc/mdx368 [DOI] [PubMed] [Google Scholar]
  72. Mould DR, Sweeney KR (2007) The pharmacokinetics and pharmacodynamics of monoclonal antibodies—mechanistic modeling applied to drug development. Curr Opin Drug Discov Devel 10(1):84–96 [PubMed] [Google Scholar]
  73. Napolitano S, Brancaccio G, Argenziano G, Martinelli E, Morgillo F, Ciardiello F et al (2018) It is finally time for adjuvant therapy in melanoma. Cancer Treat Rev 69:101–111. 10.1016/j.ctrv.2018.06.003 [DOI] [PubMed] [Google Scholar]
  74. Nathan PD, Gaunt P, Wheatley K et al (2016) UKMCC-01: A phase II study of pazopanib (PAZ) in metastatic Merkel cell carcinoma. J Clin Oncol 34:9542s. 10.1200/JCO.2016.34.15_suppl.9542 [Google Scholar]
  75. Nghiem PT, Bhatia S, Lipson EJ, Kudchadkar RR, Miller NJ, Annamalai L et al (2016) PD-1 blockade with pembrolizumab in advanced Merkel-cell carcinoma. N Engl J Med 374:2542–2552. 10.1056/NEJMoa1603702 [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Nghiem P, Kaufman HL, Bharmal M, Mahnke L, Phatak H, Becker JC (2017) Systematic literature review of efficacy, safety and tolerability outcomes of chemotherapy regimens in patients with metastatic Merkel cell carcinoma. Future Oncol 13:1263–1279. 10.2217/fon-2017-0072 [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Nghiem P, Bhatia S, Lipson EJ, Sharfman WH, Kudchadkar RR, Friedlander PA et al (2018) Durable tumor regression and overall survival (OS) in patients with advanced Merkel cell carcinoma (aMCC) receiving pembrolizumab as first-line therapy. 2018 ASCO Annual Meeting. J Clin Oncol 36(15 suppl):9506 [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. OPDIVO (nivolumab) (2018) Summary of product characteristics. https://www.ema.europa.eu/documents/product-information/opdivo-epar-product-information_en.pdf. Accessed 18 Oct 2018]
  79. Orlov S, Salari F, Kashat L, Walfish PG (2015) Induction of painless thyroiditis in patients receiving programmed death 1 receptor immunotherapy for metastatic malignancies. J Clin Endocrinol Metab 100:1738–1741. 10.1210/jc.2014-4560 [DOI] [PubMed] [Google Scholar]
  80. Parker JJ, Jones JC, Strober S, Knox SJ et al (2013) Characterization of direct radiation-induced immune function and molecular signaling changes in an antigen presenting cell line. Clin Immunol 148:44–55. 10.1016/j.clim.2013.03.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Paulson KG, Tegeder A, Willmes C, Iyer JG, Afanasiev OK, Schrama D et al (2014) Downregulation of MHC-I expression is prevalent but reversible in Merkel cell carcinoma. Cancer Immunol Res 2(11):1071–1079. 10.1158/2326-6066 [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Paulson KG, Perdicchio M, Kulikauskas R, Wagener F, Church CD, Bui K et al (2017) Augmentation of adoptive T-cell therapy for Merkel cell carcinoma with avelumab. J Clin Oncol 35:15s (suppl. abstr 3044) [Google Scholar]
  83. Peng J, Hamanishi J, Matsumura N, Abiko K, Murat K, Baba T et al (2015) Chemotherapy induces programmed cell death-ligand 1 overexpression via the nuclear factor-kappaB to foster an immunosuppressive tumor microenvironment in ovarian cancer. Cancer Res 75(23):5034–5045. 10.1158/0008-5472.can-14-3098 [DOI] [PubMed] [Google Scholar]
  84. Postow MA (2015a) Managing immune checkpoint-blocking antibody side effects. Am Soc Clin Oncol Educ Book. 10.14694/EdBook_AM.2015.35.76 [DOI] [PubMed] [Google Scholar]
  85. Postow MA, Callahan MK, Wolchok JD (2015b) Immune checkpoint blockade in cancer therapy. J Clin Oncol 33:1974–1982. 10.1200/JCO.2014.59.4358 [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Prewett SL, Ajithkumar T (2015) Merkel cell carcinoma: current management and controversies. Clin Oncol (R Coll Radiol) 27:436–444. 10.1016/j.clon.2015.04.007 [DOI] [PubMed] [Google Scholar]
  87. Rabinowits G, Lezcano C, Catalano PJ, McHugh P, Becker H, Reilly MM et al (2018) Cabozantinib in patients with advanced Merkel cell carcinoma. Oncologist 23(7):814–821. 10.1634/theoncologist.2017-0552 [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Rabinowitz G (2017) Is this the end of cytotoxic chemotherapy in Merkel cell carcinoma? OncoTargets Ther 10:4803–4807. 10.2147/OTT.S126640 [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Ritter C, Fan K, Paulson KG, Nghiem P, Schrama D, Becker JC (2016) Reversal of epigenetic silencing of MHC class I chain-related protein A and B improves immune recognition of Merkel cell carcinoma. Sci Rep 6:21678. 10.1038/srep21678 [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Robert C, Schachter J, Long G, Arance A, Grob JJ, Mortier L et al (2015) Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med 372(26):2521–2532. 10.1056/NEJMoa1503093 [DOI] [PubMed] [Google Scholar]
  91. Robert C, Ribas A, Hamid O, Daud A, Wolchok JD, Joshua AM et al (2016) Three year overall survival for patients with advanced melanoma treated with pembrolizumab in KEYNOTE-001. J Clin Oncol 34:9503 [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Rojkó L, Reiniger L, Téglási V, Fábián K, Pipek O, Vágvölgyi A et al (2018) Chemotherapy treatment is associated with altered PD-L1 expression in lung cancer patients. J Cancer Res Clin Oncol 144:1219–1226. 10.1007/s00432-018-2642-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Rosenberg JE, Hoffman-Censits J, Powles T, van der Heijden MS, Balar AV, Necchi A et al (2016) Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet 387(10031):1909–1920. 10.1016/S0140-6736(16)00561-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Rossi C, Gilhodes J, Maerevoet M, Herbaux C, Morschhauser F, Brice P et al (2018) Efficacy of chemotherapy or chemo-anti-PD-1 combination after failed anti-PD-1 therapy for relapsed and refractory Hodgkin lymphoma: a series from Lysa centers. Am J Hematol. 10.1002/ajh.25154 [DOI] [PubMed] [Google Scholar]
  95. Saleh K, Khalifeh-Saleh N, Kourie HR (2018) Is it possible to rechallenge with PD-1/PD-L1 inhibitors after progression? Immunotherapy 10(5):345–347. 10.2217/imt-2017-0180 [DOI] [PubMed] [Google Scholar]
  96. Samlowski WE, Moon J, Tuthill RJ, Heinrich MC, Balzer-Haas NS, Merl SA, DeConti RC et al (2010) A phase II trial of imatinib mesylate in merkel cell carcinoma (neuroendocrine carcinoma of the skin): a Southwest Oncology Group study (S0331). Am J Clin Oncol 33(5):495–499. 10.1097/COC.0b013e3181b9cf04 [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Schadendorf D, Lebbé C, Zur Hausen A, Avril MF, Hariharan S, Bharmal M et al (2017) Merkel cell carcinoma: Epidemiology, prognosis, therapy and unmet medical needs. Eur J Cancer 71:53–69. 10.1016/j.ejca.2016.10.022 [DOI] [PubMed] [Google Scholar]
  98. Segal NH, He AR, Doi T, Levy R, Bhatia S, Pishvaian MJ et al (2018) Phase I study of single-agent utomilumab (PF-05082566), a 4-1BB/CD137 agonist, in patients with advanced cancer. Clin Cancer Res 24(8):1816–1823. 10.1158/1078-0432.CCR-17-1922 [DOI] [PubMed] [Google Scholar]
  99. Sharabi AB, Lim M, DeWeese TL, Drake CG et al (2015) Radiation and checkpoint blockade immunotherapy: radiosensitisation and potential mechanisms of synergy. Lancet Oncol 16:498–509. 10.1016/S1470-2045(15)00007-8 [DOI] [PubMed] [Google Scholar]
  100. Shaverdian N, Lisberg AE, Bornazyan K, Veruttipong D, Goldman JW, Formenti SC et al (2017) Previous radiotherapy and the clinical activity and toxicity of pembrolizumab in the treatment of non-small-cell lung cancer: a secondary analysis of the KEYNOTE-001 phase 1 trial. Lancet Oncol 18:895–903. 10.1016/S1470-2045(17)30380-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Sheng J, Fang W, Yu J, Chen N, Zhan J, Ma Y et al (2016) Expression of programmed death ligand-1 on tumor cells varies pre and post chemotherapy in non-small cell lung cancer. Sci Rep 6:20090. 10.1038/srep20090 [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Shiver MB, Mahmoud F, Gao L (2015) Response to idelalisib in a patient with stage IV Merkel-cell carcinoma. N Engl J Med 373(16):1580–1582. 10.1056/NEJMc1507446 [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Tadmor T, Aviv A, Polliack A (2011) Merkel cell carcinoma, chronic lymphocytic leukemia and other lymphoproliferative disorders: an old bond with possible new viral ties. Ann Oncol 22:250–256. 10.1093/annonc/mdq308 [DOI] [PubMed] [Google Scholar]
  104. Tarabadkar ES, Thomas H, Blom A, Parvathaneni U, Olencki T, Nghiem P et al (2018) Clinical benefit from tyrosine kinase inhibitors in metastatic merkel cell carcinoma: a case series of 5 patients. Am J Case Rep 19:505–511. 10.12659/AJCR.908649 [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Taralli S, Sollini M, Milella M, Perotti G, Filice A, Menga M et al (2018)) 18F-FDG and 68 Ga-somatostatin analogs PET/CT in patients with Merkel cell carcinoma: a comparison study. EJNMMI Res. 10.1186/s13550-018-0423-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Tarhini AA, Edington H, Butterfield LH, Lin Y, Shuai Y, Tawbi H et al (2014) Immune monitoring of the circulation and the tumor microenvironment in patients with regionally advanced melanoma receiving neoadjuvant ipilimumab. PLoS One 9:e87705. 10.1371/journal.pone.0087705 [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Terawaki S, Tanaka Y, Nagakura T, Hayashi T, Shibayama S, Muroi K et al (2007) Specific and high-affinity binding of tetramerized PD-L1 extracellular domain to PD-1-expressing cells: possible application to enhance T cell function. Int Immunol 19(7):881–890 [DOI] [PubMed] [Google Scholar]
  108. Terheyden P, Becker JC (2017) New developments in the biology and the treatment of metastatic Merkel cell carcinoma. Curr Opin Oncol. 10.1097/CCO.0000000000000363 [DOI] [PubMed] [Google Scholar]
  109. Tilling T, Moll I (2012) Which are the cells of origin in merkel cell carcinoma? J Skin Cancer 2012:680410. 10.1155/2012/680410 [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Toker C (1972) Trabecular carcinoma of the skin. Arch Dermatol 105:107–110. 10.1001/archderm.1972.01620040075020 [PubMed] [Google Scholar]
  111. Topalian SL, Bhatia S, Kudchadkar RR, Amin A, Sharfman WH, Lebbe C et al (2018) Nivolumab (Nivo) as neoadjuvant therapy in patients with resectable Merkel cell carcinoma (MCC) in CheckMate 358. 2018 ASCO Annual Meeting. J Clin Oncol 36(15 suppl):9505 [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Uchi H (2018) Merkel Cell Carcinoma: An Update and Immunotherapy. Front Oncol 8:48. 10.3389/fonc.2018.00048. (eCollection 2018) [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Villasboas JC, Ansell SM, Witzig TE (2016) Targeting the PD-1 pathway in patients with relapsed classic Hodgkin lymphoma following allogeneic stem cell transplant is safe and effective. Oncotarget 7(11):13260–13264. 10.18632/oncotarget.7177 [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Waldherr C, Herrmann R, Maecke H, Mueller-Brand J, Pless M (2004) Successful targeted radiotherapy with 90Y-DOTATOC in a patient with Merkel cell carcinoma. A case report. Oncology 66(2):160–163. 10.1159/000077443 [DOI] [PubMed] [Google Scholar]
  115. Weber JS, Dummer R, de Pril V, Lebbé C, Hodi FS, MDX010-20 Investigators (2013) Patterns of onset and resolution of immune-related adverse events of special interest with ipilimumab: detailed safety analysis from a phase 3 trial in patients with advanced melanoma. Cancer 119:1675–1682. 10.1002/cncr.27969 [DOI] [PubMed] [Google Scholar]
  116. Weber J, Mandala M, Del Vecchio M, Gogas HJ, Arance AM, Cowey CL et al (2017) Adjuvant Nivolumab versus ipilimumab in resected stage III or IV melanoma. N Engl J Med 377:1824–1835. 10.1056/NEJMoa1709030 [DOI] [PubMed] [Google Scholar]
  117. Wehkamp U, Stern S, Krüger S, Hauschild A, Röcken C, Egberts F (2017) Tropomyosin receptor kinase a expression on merkel cell carcinoma cells. JAMA Dermatol 153(11):1166–1169. 10.1001/jamadermatol.2017.2495 [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Yared JA, Hardy N, Singh Z, Hajj S, Badros AZ, Kocoglu M et al (2016) Major clinical response to nivolumab in relapsed/refractory Hodgkin lymphoma after allogeneic stem cell transplantation. Bone Marrow Transpl 51(6):850–852. 10.1038/bmt.2015.346 [DOI] [PubMed] [Google Scholar]
  119. Zhang P, Su DM, Liang M, Fu J (2008) Chemopreventive agents induce programmed death-1-ligand 1 (PD-L1) surface expression in breast cancer cells and promote PD-L1-mediated T cell apoptosis. Mol Immunol 45(5):1470–1476. 10.1016/j.molimm.2007.08.013 [DOI] [PubMed] [Google Scholar]
  120. Zhang P, Ma Y, Lv C, Huang M, Li M, Dong B et al (2016) Upregulation of programmed cell death ligand 1 promotes resistance response in non-small-cell lung cancer patients treated with neo-adjuvant chemotherapy. Cancer Sci 107(11):1563–1571. 10.1111/cas.13072 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Cancer Research and Clinical Oncology are provided here courtesy of Springer

RESOURCES