Abstract
Merkel cell carcinoma (MCC) is a rare skin malignancy that is a paradigm cancer for solid tumor immunotherapy. MCCs associated with Merkel cell polyomavirus (virus-positive MCC [VP-MCC]) or chronic UV exposure (virus-negative MCC [VN-MCC]) are anti–PD(L)1 responsive, despite VP-MCC’s low mutational burden. This suggests that antigen quality, not merely mutation quantity, dictates immunotherapy responsiveness, and cell-based therapies targeting optimal antigens may be effective. Despite VP-MCC’s antigenic homogeneity, diverse T-cell infiltration patterns are observed, implying microenvironment plasticity and multifactorial contributions to immune recognition. Moreover, VP-MCC exemplifies how antitumor adaptive immunity can provide tumor burden biomarkers for early detection and disease monitoring.
Introduction
Merkel cell carcinoma (MCC) is a rare skin cancer with increasing incidence. It disproportionally affects elderly and immunocompromised persons, indicating that immune suppression favors disease progression (Albores-Saavedra et al., 2010; Penn and First, 1999). Risk factors include UV light exposure and fair skin, similar to other skin malignancies and consistent with UV-derived mutations driving oncogenesis (Albores-Saavedra et al., 2010; Heath et al., 2008; Wong et al., 2015). In 2008, clonal integration of the Merkel cell polyomavirus (MCPyV) revealed a second oncogenic driver (Feng et al., 2008), subdividing MCC into virus-positive MCC (VP-MCC) (~80% of cases in the United States), and UV-associated MCCs (virus-negative MCC [VN-MCC]) (Figure 1) (Becker et al., 2009; Feng et al., 2008; Kassem et al., 2008). VP-MCCs are driven by obligate expression of conserved MCPyV-derived oncoproteins and large and small T antigen, with few additional genomic mutations (Goh et al., 2016; Houben et al., 2010; Knepper et al., 2019). This makes VP-MCC genetically quite similar across patients and considerably more homogeneous than most cancers. Uniform (VP-MCC) and diverse (VN-MCC) variants of the same cancer make MCC a natural model for studying cancere–immune interactions.
Immune checkpoint blockade (ICB) (Ishida et al., 1992; Leach et al., 1996) of the PD-1 axis has shown widespread clinical benefit across cancer types (Brahmer et al., 2012; Galanina et al., 2018; Herbst et al., 2014; Nghiem et al., 2016; Yarchoan et al., 2017). ICB improves outcomes for approximately 50% of patients with MCC—one of the highest solid tumor response rates (Becker et al., 2017; Paulson et al., 2019)—and National Comprehensive Cancer Network guidelines now recommend ICB therapy as first-line treatment for metastatic MCC (Bichakjian et al., 2018). MCC has a relatively even distribution of ICB responders (30–50%), nonresponders (25–30%), and escapees (25–30%) (Kaufman et al., 2016; Nghiem et al., 2016) across both pathogenic pathways, again rendering MCC a prototype cancer.
In this review, we use MCC as a paradigm cancer for understanding ICB response and resistance. We first explore how MCC—with dual pathogenic pathways and even distribution of responders, nonresponders, and escapees—can be used to model molecular mechanisms by assessing epitope quality and immune cell infiltrate patterns. We then discuss how cell-based immunotherapies for MCC provide a pertinent model for refining immunotherapy development. Finally, with MCC as an example, we consider strategies utilizing immune cells for disease detection.
Highly immunogenic tumor-specific epitopes facilitate ICB response
Although ICB has revolutionized cancer therapy, outcomes vary and, currently, cannot be accurately predicted. For example, higher expression of the PD-1 ligand CD274, commonly denoted PD-L1, on tumor or tumor-associated immune cells correlates with improved response but does not reliably separate responders from nonresponders (Herbst et al., 2014). For VP-MCC, VN-MCC, and other immunogenic solid tumors (e.g., HIV-associated Kaposi Sarcoma [HIV-KS]), post-ICB remissions are observed even with undetectable PD-L1 expression. A more widely used predictor of ICB response is median tumor mutational burden (TMB), which positively correlates with ICB response (Yarchoan et al., 2017). Not all cancers follow this correlation, emphasizing the need to precisely understand mechanisms governing response and resistance so that accurate predictors can be identified (Galanina et al., 2018; Lipson et al., 2013). Deciphering immune interactions is challenging, because tumor mutations vary between tumors and different T-cell clones can recognize identical mutations when presented by HLA. Uniform (VP-MCC) and diverse (VN-MCC) variants provide a uniquely suitable platform in which to investigate this heterogeneity in the context of both high and low TMB.
Tumor-specific antigen quality is associated with response to PD-1 axis blockade.
The positive correlation between TMB and ICB response implies that heavily mutated tumors are more immunogenic and subsequently more likely to respond (Cristescu et al., 2018; Le et al., 2017, 2015; Rizvi et al., 2015; Yarchoan et al., 2017). The molecular basis of this observation remains unclear, particularly with regard to a tumor’s immunopeptidome, that is, the sum of presented epitopes (intracellularly derived peptides presented on HLA) recognized by cytotoxic T cells that subsequently mount an immune response. Is this correlation between TMB and ICB response driven by TMB-high tumors having numerous, minimally immunogenic epitopes or a few, highly immunogenic epitopes? MCC’s dual etiology in conjunction with other virus-associated cancers provides an excellent opportunity to address this question and suggest that epitope quality is consequential.
VN-MCC has a high TMB (median, 63.1 mutations per Megabase [MB]; range, 31–133), consistent with other UV-associated skin cancers (Knepper et al., 2019). Conversely, VP-MCC has an exceptionally low TMB (median, 1.2 mutations per MB; range, 0.0–2.6) and few other genomic aberrations (Goh et al., 2016; Knepper et al., 2019; Paulson et al., 2009). VN-MCC and VP-MCC have similar overall response rates (ORRs) (44–54%) to anti-PD-1 blockade. For VN-MCC, this high ORR is predicted by median TMB (Goh et al., 2016; Goodman et al., 2019, 2017; Harms et al., 2015; Yarchoan et al., 2019, 2017). VP-MCC has a substantially better ORR than TMB alone would predict (Goh et al., 2016; Knepper et al., 2019; Nghiem et al., 2019, 2016). Although VP-MCC has low TMB and subsequently few predicted neoepitopes, it does contain MCPyV viral proteins (Berry et al., 2019; Iyer et al., 2011) that generate highly immunogenic epitopes. Its viral protein sequence is foreign, and thus MCPyV-specific T cells recognize its HLA-presented epitopes generally with higher avidity or ability to efficiently kill VP-MCC cells, because these T cells have not undergone thymic-negative selection (Croft et al., 2019). Furthermore, the MCPyV T antigen locus codes for proteins under 900 amino acids in length (Spurgeon and Lambert, 2013). This small, finite, viral epitope repertoire and high ORR suggests that limited but highly immunogenic epitopes may be sufficient for favorable responses.
To explore if tumor-specific antigen quality influenced ICB ORRs, we analyzed virus- and nonvirus-associated cancers separately. As a starting point, Dr. Mark Yarchoan kindly shared his published data set comparing PD-1 ORR and TMB across several cancers (Yarchoan et al., 2017). Five virus-associated cancers were identified—VP-MCC, HIV-KS (HHV-8 driven), virus-positive head and neck (VP-H&N), cervical, and anal cancers (latter three, human papillomavirus driven). The specific etiology was not available for HIV-KS, cervical, or anal cancers; these are >90% virus-driven (Chang et al., 1994; Joseph et al., 2008; Walboomers et al., 1999). However, a skewing of the associated response rates could not be formally excluded. Because virus-associated etiology was not available for VP-MCC and VP-H&N in the Yarchaon dataset, these data were removed and replaced with data from studies that explicitly divided patients based on cancer etiology (Chow et al., 2016; Harms et al., 2015; Nghiem et al., 2019; Seiwert et al., 2015; Yarchoan et al., 2017). Log transformations, linear regressions, and tests of linear combinations of regression coefficients were performed using R.
When virus-associated cancers were removed from the data set, leaving only nonvirus-associated cancers, the correlation between TMB and ORR increased (from 0.488 to 0.849; n = 25). In contrast, the virus-associated cancers (n = 5) showed a negative correlation (R2 = −0.619), suggesting that TMB may not be an adequate predictor for all cancers (Figure 2). Thus, separating virus-positive and virus-negative cancers will be critical moving forward in deepening our understanding of TMB and ORR.
All virus-associated cancers analyzed exhibited higher ORR than TMB alone might predict. For the same TMB, virus-associated cancers have an average of 25.3% higher ORR than nonvirus cancers (95% confidence interval = 2.4–48.2; P = 0.03). Nonvirus-associated cancers require 30 times greater TMB to elicit responses similar to those in virus-associated cancers (Chow et al., 2016; Goh et al., 2016; Harms et al., 2015; Nghiem et al., 2019; Seiwert et al., 2015; Yarchoan et al., 2017). On average, nonvirus-associated cancers (n = 25) had a TMB of 11.6 and ORR of 17.4, whereas virus-associated cancers (n = 5) had a TMB of 5.7 and ORR of 39.4. Although it cannot be deduced that multiple less immunogenic neoepitopes could also be effective in tumors with high TMB, these data suggest that a small number of highly immunogenic epitopes can elicit ICB responses relative to a large number of less immunogenic epitopes.
Oligoclonal expanded tumor-infiltrating lymphocytes suggest limited quantity of immunogenic epitopes.
In melanoma, a virus-negative cancer with high TMB, very few tumor-derived epitopes are neoantigens (<0.1%) or unique tumor-associated antigens (1.7–3%). Analysis of tumor-infiltrating lymphocytes (TILs) from these same melanomas showed expansion of T-cell clones specific to the few neoepitopes and unique tumor-associated epitopes, again implying that limited but potent epitopes can drive T-cell responses (Kalaora et al., 2018). Furthermore, high-throughput genomic and proteomic methods have shown that relatively few of the total possible neoepitopes can mediate tumor rejection (Ebrahimi-Nik et al., 2019; Yadav et al., 2014). Identifying these needle in the haystack epitopes driving immunogenic tumor rejection will be critical for developing broadly applicable vaccination or engineered cell therapies. VP-MCC offers a unique advantage for piloting such therapies owing to a small antigenic space (MCPyV T antigens) to test approaches, and shared, tumor-specific antigens across patients assure broad applicability.
Tumor-specific T-cell response patterns are plastic and independent of tumor-associated antigen expression.
Similar to TMB, TIL patterns can partially predict ICB response.(Chen and Mellman, 2017; Cristescu et al., 2018; Gruosso et al., 2019) Three distinct patterns described by Mellman and others are observed in MCC (Paulson et al., 2011) and other solid tumors (Chen and Mellman, 2017) (Figure 1b): (i) T cell desert—no T cells detected in or near tumor. These tumors rarely respond to ICB and are common in relapsed metastases (Chen and Mellman, 2017; Knepper et al., 2019; Paulson et al., 2018); (ii) T cell exclusion— T cells concentrate on edges but fail to infiltrate the tumor because of a physical and/or chemical barrier. These tumors sometimes respond to ICB therapy (Giraldo et al., 2018; Knepper et al., 2019; Paulson et al., 2018); and (iii) T cell inflamed—T cells observed throughout the tumor. This phenotype is frequently seen during response and is typically associated with improved prognosis (Chen and Mellman, 2017; Gruosso et al., 2019; Herbst et al., 2014; Paulson et al., 2018).
Determining whether TIL patterns are a consequence of the microenvironment or variations of tumor epitope immunogenicity is exceptionally challenging. Indeed, large-scale investigations of these features have been unfruitful (Tamborero et al., 2018; Thorsson et al., 2018). Similarly, allelic diversity in presentation molecules, particularly class I HLA, can influence immunogenicity, further complicating studies (Crux and Elahi, 2017; Goulder and Watkins, 2008; Kiepiela et al., 2004). The field does recognize that immunogenic antigens (e.g., viral proteins) play a critical role in activating lymphocytes and recruiting T cells to tumors (Gameiro et al., 2018; Miller et al., 2018). Here, VP-MCC provides an informative natural system in which obligate viral oncoproteins and low TMB imply relatively homogeneous immunogenicity. Therefore, in this system, TIL pattern differences are not directly related to the tumor antigen itself (MCPyV) but instead associated with microenvironment fluctuations or HLA polymorphisms rendering the same antigen (MCPyV) more or less immunogenic. Surprisingly, all three TIL patterns have been recorded in the same patient with VP-MCC at different stages of response and resistance to T-cell immunotherapy (Paulson et al., 2018), suggesting that complex microenvironment factors dictate T-cell infiltration. Moving forward, sophisticated systems analyses will be necessary to precisely identify which factors drive or inhibit T-cell infiltration.
MCC serves as a model cancer in which to understand factors mediating immunotherapy response and resistance
Many patients’ tumors progress after starting ICB treatment (primary resistance) or after a period of initial response (acquired resistance) (Sharma et al., 2017). For MCC, ~30% of initial responders progress by 2 years after ICB initiation (Nghiem et al., 2019).
Primary resistance is generally a direct consequence of a patient’s T cells being unable to respond to tumor, rendering ICB ineffective (Pos et al., 2014; Sharma et al., 2017). This may be driven by a number of factors: tumor lacking target antigen(s) (Gubin et al., 2014), dysfunctional antigen presentation (Marincola et al., 2000; Sucker et al., 2014), advanced or irreversible tumor-specific T-cell dysfunction, and/or lack of tumor-specific T cells. Primary-resistant tumors presumably experience little immunogenic pressure from ICB, making the tumor unlikely to undergo major shifts; therefore, primary-resistant tumors are not often utilized in studying escape mechanisms. Tumors with initial response followed by acquired resistance offer an opportunity to rigorously examine mechanisms of resistance. Identifying the root cause of acquired resistance remains challenging, as multiple contributors can produce the same clinical phenotype.
Immune escape mechanisms fit into two broad categories, immune cell intrinsic or tumor intrinsic. Immune cell intrinsic mechanisms include changes to innate immune cells, such as reduced neutrophil chemotaxis (Akhbari et al., 2018) and toll-like receptor 9 downregulation (Shahzad et al., 2013), and changes to adaptive immune cells, such as T-cell exhaustion (PD-1, CTLA-4, TIM3) (Afanasiev et al., 2013b) and T-cell migration and infiltration patterns (Afanasiev et al., 2013a; Clark et al., 2008; Dowlatshahi et al., 2013; Feldmeyer et al., 2016; Sihto et al., 2012). Tumor intrinsic changes include HLA loss (Paulson et al., 2018, 2014b; Ritter et al., 2016) and proliferation of a preexisting, resistant clone (Sharma et al., 2017).
Precisely deciphering if resistance is immune cell intrinsic or tumor intrinsic is complex (Sharma et al., 2017). It requires simultaneously knowing which T cells can see tumor (immune cell intrinsic factor, TCR clonotypes) and what is recognized by T cells (tumor intrinsic factor, antigen and/or epitope). Often it is difficult to know both, given that discovery is codependent, that is, identifying a tumor-specific T-cell clone requires knowing which HLA-presented epitope is recognized, and vice versa. Again, VP-MCC homogeneity provides an exceptional model system.
Shared oncoprotein in VP-MCC facilitates tracking of tumor-specific T cells.
In VP-MCC, continuous expression of a known, shared viral oncoprotein MCPyV large-T antigen (LT-Ag) facilitates identification of LT-Ag–specific T cells, using fluorophore-labeled epitope:HLA tetramers (Burrows et al., 2000; Chapuis et al., 2013; Greiner et al., 2010). Tetramers tiled across LT-Ag discern MCC-specific T cells from other, quiescent T cells and can reveal epitope spread; that is, nascent immune responses against epitopes not targeted by initial immunotherapy (Paulson et al., 2017). The phenotype or TIL pattern of tumor-specific T cells can be further examined to inform molecular drivers of tumor-immune interactions and, ultimately, immunotherapy resistance.
Clinical trials using adoptively transferred MCPyV-specific T cells in VP-MCC (Chapuis et al., 2014; Paulson et al., 2018) provide an opportunity to directly study cancere–immune interactions with easily identifiable T cells recognizing a defined epitope. Briefly, T cells are isolated from patients’ peripheral blood; MCC-specific cells are expanded ex vivo against a known epitope (e.g., LT-Ag92–101), purified by specific tetramer-binding, and reinfused into the patient (Chapuis et al., 2014; Paulson et al., 2018). Transferred, polyclonal MCC-specific T cells all recognize the same epitope (e.g., LT-Ag92–101). Persistence is monitored with LT-Ag92–101 tetramers and additional markers, including PD-1, can identify functional phenotypes. Similarly, tetramers tiled across LT-Ag can elucidate antigen spread. Responding patients who received MCC-specific T cells and avelumab (anti–PD-L1) showed expansion of both MCC-specific T cells and T cells recognizing other LT-Ag epitopes (Paulson et al., 2017). Adoptive T-cell trials provide valuable details characterizing tumor-specific T cells throughout ICB therapy, insights into T celleintrinsic or tumor-specific resistance mechanisms (Paulson et al., 2018), and novel epitopes for adoptive T-cell therapy (Cheever et al., 2009; Gulley et al., 2017; Paulson et al., 2017).
MCC cell-based immunotherapies offer insight to cancer–immune interactions.
Cell-based therapies facilitate identifying, tracking, and profiling of tumor-specific immune cells. Similar to adoptive T-cell transfer, TCR-transduced T cells (TCR-T) uniformly recognize a defined epitope:HLA complex. Multiple MCPyV epitopes have been identified, and many are being considered as TCR-T targets for patients with VP-MCC, including in combination therapy trials (NCT03747484) (Gavvovidis et al., 2018; Iyer et al., 2011). Unfortunately, therapies targeting MCPyV oncoproteins are only applicable to patients with VP-MCC. To broaden the clinical scope, alternate targets are being considered, including cancer testes antigens (Dasgeb et al., 2019) and aberrantly glycosylated proteins (e.g., MUC-1) (Kurzen et al., 2003). Targeting surface proteins with chimeric antigen receptor T cells recognizing glypican-3 is also being pursued in a phase I clinical trial. Other immune cells can likewise be therapeutic. An ongoing phase 2 clinical trial uses activated NK cells and ALT-803 (IL-15) to treat patients with MCC (NCT02465957). Cell-based therapies offer naturally controlled systems with unprecedented opportunities to observe response and resistance mechanisms, cancere–immune interactions, and tumor microenvironments in vivo. Lessons learned will likely be broadly applicable, offering insights for next-generation immunotherapies.
Acquired resistance mechanisms provide rationale for therapeutic combinations.
Tumor cells can adapt to escape immune pressure. In MCC, loss or reduction of class I HLA correlates with worse prognosis, independent of immunotherapy intervention (Chowell et al., 2018; Paulson et al., 2018). Increased prevalence of cytotoxic CD8+ T cells that recognize specific epitope:HLA complexes (Rock et al., 2016) is a positive prognostic indicator (Naito et al., 1998; Paulson et al., 2014a; Shimizu et al., 2019), suggesting that CD8+ T cells play a prominent role in disease clearance. Of note, many viruses evolved specific mechanisms to evade immune recognition, including downregulation of class I HLAs (Fletcher et al., 1998; Koutsakos et al., 2019). Evasion mechanisms employed by virus-associated cancers could differ, and some may not apply to all cancer types, but understanding these evasion mechanisms will undoubtedly inform cancere–immune interactions more broadly. Helper CD4+ T cells are also emerging as integral to coordinating effective immune responses, including by producing cytokines that upregulate class I HLAs (Bhat et al., 2017; Drew et al., 1993; Phares et al., 2012; Shankaran et al., 2001). Indeed, engineered CD4+ TCR-T cells are being considered as therapeutics (Longino et al., 2019). MCC can effectively dampen CD8+ T-cell recognition by decreasing class I diversity or expression, making CD4+ T-cell employment a reasonable strategy to drive immune response in tumors with limited class I (Chowell et al., 2018; Kreiter et al., 2015; Paulson et al., 2018).
PD-1 or PD-L1 ICB-resistant tumors often show increased expression of other negative immune regulators, including TIM3, LAG3, CTLA-4, and TIGIT, suggesting that inhibiting more than one immune regulator could improve responses and/or expand the patient population benefiting from ICB (Anderson et al., 2016; Golden-Mason et al., 2009; Koyama et al., 2016; McMahan et al., 2010). Indeed, combination ICB therapy can improve clinical response (Chapuis et al., 2014; Curran et al., 2010; Paulson et al., 2017). Another combination therapy trial (NCT02584829) is testing if dual or triple ICB treatment regimens combined with cell-based immunotherapies are synergistic in MCC.
ICB therapies place selective pressure on cancers with preexisting genomic instability and heterogeneity, which can lead to resistance and/or relapse. Understanding these escape mechanisms is essential to broadening immunotherapy benefits (Mehta et al., 2018; Pai et al., 2019; Paulson et al., 2018). Targeting multiple resistance mechanisms will likely be necessary to improve outcomes and minimize late immunotherapy escape (Sharma et al., 2017). Engineered cell therapies in VP-MCC offer remarkable cancer and immune cell homogeneity. Together, they provide an unprecedented opportunity to clarify underlying mechanisms and to validate advanced therapeutic approaches.
Utilizing the immune system for more than therapy
Our immune system amplifies signals, a characteristic that can be used to monitor disease (Paulson et al., 2017, 2010). For example, antibody titers are commonly used in clinics to determine immune status (Spradling et al., 2013) and could be extended to oncology for tracking tumor burden or B-cell and T-cell clonal expansion.
MCC demonstrates tumor antigenicity that can be exploited for disease monitoring.
Foreign proteins derived from MCPyV and certain other cancer-associated antigens can induce antibody production that is measurable in peripheral blood (Kikuchi et al., 1995; Paulson et al., 2010). Circulating MCPyV T antigen-specific antibodies are rare in healthy individuals but present in most patients with VP-MCC. Oncoprotein-specific antibody titers fall during clinical response and spike before relapse, offering an easy, affordable, noninvasive, and predictive method for monitoring disease that is now included in MCC management guidelines (Bichakjian et al., 2018; Paulson et al., 2017). Similarly, in non-small cell lung cancer, circulating tumor-specific antibodies noninvasively inform disease state by differentiating benign and malignant lung nodules (Lastwika et al., 2019). Changes in antibody titers alert physicians before clinical relapse, guiding early intervention for optimal patient outcomes (Miller et al., 2018; Paulson et al., 2017).
T cells and B cells can likewise be employed to detect tumor burden and direct treatment (Chapuis et al., 2019, 2017; Grupp et al., 2013; Logan et al., 2011; Robins, 2013; Wu et al., 2012). Variable regions of B-cell receptors (BCRs) or TCRs serve as a natural barcode generated in B-cell and T-cell development. In B-cell and T-cell malignancies, BCR or TCR clonotype sequencing is used to monitor tumor progression via expansion or contraction of a cancerous lymphocytic clone (Grupp et al., 2013; Wu et al., 2012) and to detect minimal residual disease (Logan et al., 2011; Wu et al., 2012). These same sequencing methods track TCRs for monitoring persistence and efficacy of engineered T-cell therapies (Chapuis et al., 2019, 2017).
Developing antibody arrays or immune cell monitoring for additional cancers could promote early detection, direct therapy, and improve patient outcomes. Employing these methodologies to evaluate hard-to-screen cancers could replace current, highly invasive procedures and costly imaging.
Conclusions
ICB therapy success has greatly improved outcomes for patients with many previously lethal malignancies, yet many obstacles remain. Analyses revealed an unmet need to accurately predict response, which requires a deeper understanding of mechanisms governing response, resistance, and relapse. The MCC model suggests that cancer epitope quality is associated with successful ICB response. Diverse immune infiltrate patterns demonstrate microenvironment plasticity in MCC and underscore a need for predictive diagnostics. Broadening the range of validated immunotherapeutic targets will be imperative to reducing late therapy escape, as demonstrated by improved response with combination regimens in MCC. Using MCC as a paradigm cancer not only broadly informs molecular immunology and cancer immunotherapy but also supports using endogenous immune responses to monitor disease and direct treatment.
ACKNOWLEDGMENTS
This work was supported by the SITC-Merck Fellowship (KGP), Immunotherapy Integrated Research Center at Fred Hutchinson Cancer Research Center (KGP, AGC), NIH T32CA009515 (KGP), NIH T32GM095421 and NIH T32CA080416 (MCL), NIH P01 CA225517 (PTN and AGC), and Cancer Center Support Grant P30 CA015704.
CONFLICT OF INTEREST
KGP receives research funding to her previous institution from EMD Serono, Bluebird Biosciences, and SITC-Merck. AGC receives research funding to her institution from EMD Serono, Bluebird Biosciences, Juno Therapeutics, SignalOne, and Amazon. KGP and AGC hold intellectual property related to TCRs for therapy of cancer and other diseases. PTN receives research funding to his institution from EMD Serono and Bristol-Myers Squibb and has served as a paid consultant for EMD Serono, Pfizer, 4SC, and Merck Sharp & Dohme. The remaining authors state no conflict of interest.
Abbreviations:
- BCR
B-cell receptor
- HIV-KS
HIV-associated Kaposi Sarcoma
- ICB
Immune checkpoint blockade
- LT-Ag
large-T antigen
- MB
Megabase
- MCC
Merkel cell carcinoma
- MCPyV
Merkel cell polyomavirus
- ORR
overall response rate
- TCR-T
TCR-transduced T cell
- TIL
tumor-infiltrating lymphocyte
- TMB
tumor mutational burden
- VN-MCC
virus-negative Merkel cell carcinoma
- VP-H&N
virus-positive head and neck
- VP-MCC
virus-positive Merkel cell carcinoma
REFERENCES
- Afanasiev OK, Nagase K, Simonson W, Vandeven N, Blom A, Koelle DM, et al. Vascular E-selectin expression correlates with CD8 lymphocyte infiltration and improved outcome in Merkel cell carcinoma. J Invest Dermatol 2013a;133:2065–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Afanasiev OK, Yelistratova L, Miller N, Nagase K, Paulson K, Iyer JG, et al. Merkel polyomavirus-specific T cells fluctuate with Merkel cell carcinoma burden and express therapeutically targetable PD-1 and Tim-3 exhaustion markers. Clin Cancer Res 2013b;19:5351–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Akhbari P, Tobin D, Poterlowicz K, Roberts W, Boyne JR. MCV-miR-M1 targets the host-cell immune response resulting in the attenuation of neutrophil chemotaxis. J Invest Dermatol 2018;138:2343–54. [DOI] [PubMed] [Google Scholar]
- Albores-Saavedra J, Batich K, Chable-Montero F, Sagy N, Schwartz AM, Henson DE. Merkel cell carcinoma demographics, morphology, and survival based on 3870 cases: a population based study. J Cutan Pathol 2010;37:20–7. [DOI] [PubMed] [Google Scholar]
- Anderson AC, Joller N, Kuchroo VK. Lag-3, Tim-3, and TIGIT: co-inhibitory receptors with specialized functions in immune regulation. Immunity 2016;44:989–1004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Becker JC, Houben R, Ugurel S, Trefzer U, Pföhler C, Schrama D. MC polyomavirus is frequently present in Merkel cell carcinoma of European patients. J Invest Dermatol 2009;129:248–50. [DOI] [PubMed] [Google Scholar]
- Becker JC, Stang A, DeCaprio JA, Cerroni L, Lebbé C, Veness M, et al. Merkel cell carcinoma. Nat Rev Dis Primers 2017;3:17077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berry JG, Goodman DM, Coller RJ, Agrawal R, Kuo DZ, Cohen E, et al. Association of home respiratory equipment and supply use with health care resource utilization in children. J Pediatr 2019;207:169–75.e2. [DOI] [PubMed] [Google Scholar]
- Bhat P, Leggatt G, Waterhouse N, Frazer IH. Interferon-gamma derived from cytotoxic lymphocytes directly enhances their motility and cytotoxicity. Cell Death Dis 2017;8:e2836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bichakjian CK, Olencki T, Aasi SZ, Alam M, Andersen JS, Blitzblau R, et al. Merkel cell carcinoma, version 1.2018, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw 2018;16:742–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 2012;366:2455–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burrows SR, Kienzle N, Winterhalter A, Bharadwaj M, Altman JD, Brooks A. Peptide-MHC class I tetrameric complexes display exquisite ligand specificity. J Immunol 2000;165:6229–34. [DOI] [PubMed] [Google Scholar]
- Chang Y, Cesarman E, Pessin MS, Lee F, Culpepper J, Knowles DM, et al. Identification of herpesvirus-like DNA sequences in AIDS-associated Kaposi’s sarcoma. Science 1994;266:1865–9. [DOI] [PubMed] [Google Scholar]
- Chapuis AG, Afanasiev OK, Iyer JG, Paulson KG, Parvathaneni U, Hwang JH, et al. 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 2014;2:27–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chapuis AG, Desmarais C, Emerson R, Schmitt TM, Shibuya K, Lai I, et al. Tracking the fate and origin of clinically relevant adoptively transferred CD8+ T cells in vivo. Sci Immunol 2017;2:eaal2568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chapuis AG, Egan DN, Bar M, Schmitt TM, McAfee MS, Paulson KG, et al. T cell receptor gene therapy targeting WT1 prevents acute myeloid leukemia relapse post-transplant. Nat Med 2019;25:1064–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chapuis AG, Ragnarsson GB, Nguyen HN, Chaney CN, Pufnock JS, Schmitt TM, et al. Transferred WT1-reactive CD8+ T cells can mediate antileukemic activity and persist in post-transplant patients. Sci Transl Med 2013;5:174ra27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheever MA, Allison JP, Ferris AS, Finn OJ, Hastings BM, Hecht TT, et al. The prioritization of cancer antigens: a National Cancer Institute pilot project for the acceleration of translational research. Clin Cancer Res 2009;15: 5323–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature 2017;541:321–30. [DOI] [PubMed] [Google Scholar]
- Chow LQM, Haddad R, Gupta S, Mahipal A, Mehra R, Tahara M, et al. Antitumor activity of pembrolizumab in biomarker-unselected patients with recurrent and/or metastatic head and neck squamous cell carcinoma: results from the phase Ib KEYNOTE-012 expansion cohort. J Clin Oncol 2016;34:3838–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chowell D, Morris LGT, Grigg CM, Weber JK, Samstein RM, Makarov V, et al. Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science 2018;359:582–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clark RA, Huang SJ, Murphy GF, Mollet IG, Hijnen D, Muthukuru M, et al. Human squamous cell carcinomas evade the immune response by downregulation of vascular E-selectin and recruitment of regulatory T cells. J Exp Med 2008;205:2221–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cristescu R, Mogg R, Ayers M, Albright A, Murphy E, Yearley J, et al. Pantumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science 2018;362:eaar3593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Croft NP, Smith SA, Pickering J, Sidney J, Peters B, Faridi P, et al. Most viral peptides displayed by class I MHC on infected cells are immunogenic. Proc Natl Acad Sci USA 2019;116:3112–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crux NB, Elahi S. Human leukocyte antigen (HLA) and immune regulation: how do classical and non-classical HLA alleles modulate immune response to human immunodeficiency virus and hepatitis C virus infections? Front Immunol 2017;8:832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Curran MA, Montalvo W, Yagita H, Allison JP. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci USA 2010;107: 4275–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dasgeb B, Mehregan D, Ring C, Nartker N, Brownell I. Cancer-testis antigens as biomarkers for Merkel cell carcinoma: pitfalls and opportunities. J Cutan Pathol 2019;46:748–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dowlatshahi M, Huang V, Gehad AE, Jiang Y, Calarese A, Teague JE, et al. Tumor-specific T cells in human Merkel cell carcinomas: a possible role for Tregs and T-cell exhaustion in reducing T-cell responses. J Invest Dermatol 2013;133:1879–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Drew PD, Lonergan M, Goldstein ME, Lampson LA, Ozato K, McFarlin DE. Regulation of MHC class I and beta 2-microglobulin gene expression in human neuronal cells. Factor binding to conserved cis-acting regulatory sequences correlates with expression of the genes. J Immunol 1993;150: 3300–10. [PubMed] [Google Scholar]
- Ebrahimi-Nik H, Michaux J, Corwin WL, Keller GL, Shcheglova T, Pak H, et al. Mass spectrometry driven exploration reveals nuances of neoepitopedriven tumor rejection. JCI Insight 2019;5:e129152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feldmeyer L, Hudgens CW, Ray-Lyons G, Nagarajan P, Aung PP, Curry JL, et al. Density, distribution, and composition of immune infiltrates correlate with survival in Merkel cell carcinoma. Clin Cancer Res 2016;22: 5553–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feng H, Shuda M, Chang Y, Moore PS. Clonal integration of a polyomavirus in human Merkel cell carcinoma. Science 2008;319:1096–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fletcher JM, Prentice HG, Grundy JE. Natural killer cell lysis of cytomegalovirus (CMV)-infected cells correlates with virally induced changes in cell surface lymphocyte function-associated antigen-3 (LFA-3) expression and not with the CMV-induced down-regulation of cell surface class I HLA. J Immunol 1998;161:2365–74. [PubMed] [Google Scholar]
- Galanina N, Goodman AM, Cohen PR, Frampton GM, Kurzrock R. Successful treatment of HIV-associated Kaposi sarcoma with immune checkpoint blockade. Cancer Immunol Res 2018;6:1129–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gameiro SF, Ghasemi F, Barrett JW, Koropatnick J, Nichols AC, Mymryk JS, et al. Treatment-naive HPV+ head and neck cancers display a T-cell-inflamed phenotype distinct from their HPV- counterparts that has implications for immunotherapy. Oncoimmunology 2018;7:e1498439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. Clin Cancer Res 2018;24:3644–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. J Immunother Cancer 2018;6:99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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;7:3403–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Golden-Mason L, Palmer BE, Kassam N, Townshend-Bulson L, Livingston S, McMahon BJ, et al. Negative immune regulator Tim-3 is overexpressed on T cells in hepatitis C virus infection and its blockade rescues dysfunctional CD4+ and CD8+ T cells. J Virol 2009;83:9122–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goodman AM, Kato S, Bazhenova L, Patel SP, Frampton GM, Miller V, et al. Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol Cancer Ther 2017;16:2598–608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goodman AM, Sokol ES, Frampton GM, Lippman SM, Kurzrock R. Microsatellite-stable tumors with high mutational burden benefit from immunotherapy. Cancer Immunol Res 2019;7:1570–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goulder PJ, Watkins DI. Impact of MHC class I diversity on immune control of immunodeficiency virus replication. Nat Rev Immunol 2008;8:619–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Greiner J, Schmitt A, Giannopoulos K, Rojewski MT, Götz M, Funk I, et al. High-dose RHAMM-R3 peptide vaccination for patients with acute myeloid leukemia, myelodysplastic syndrome and multiple myeloma. Haematologica 2010;95(7):1191–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gruosso T, Gigoux M, Manem VSK, Bertos N, Zuo D, Perlitch I, et al. Spatially distinct tumor immune microenvironments stratify triple-negative breast cancers. J Clin Invest 2019;129:1785–800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med 2013;368:1509–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gubin MM, Zhang X, Schuster H, Caron E, Ward JP, Noguchi T, et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 2014;515:577–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gulley JL, Madan RA, Pachynski R, Mulders P, Sheikh NA, Trager J, et al. Role of antigen spread and distinctive characteristics of immunotherapy in cancer treatment. J Natl Cancer Inst 2017;109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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 Res 2015;75:3720–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heath M, Jaimes N, Lemos B, Mostaghimi A, Wang LC, Peñas PF, et al. Clinical characteristics of Merkel cell carcinoma at diagnosis in 195 patients: the AEIOU features. J Am Acad Dermatol 2008;58: 375–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014;515:563–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Houben R, Shuda M, Weinkam R, Schrama D, Feng H, Chang Y, et al. Merkel cell polyomavirus-infected Merkel cell carcinoma cells require expression of viral T antigens. J Virol 2010;84:7064–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J 1992;11:3887–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. Clin Cancer Res 2011;17: 6671–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Joseph DA, Miller JW, Wu X, Chen VW, Morris CR, Goodman MT, et al. Understanding the burden of human papillomavirus-associated anal cancers in the US. Cancer 2008;113(Suppl.):2892–900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalaora S, Wolf Y, Feferman T, Barnea E, Greenstein E, Reshef D, et al. Combined analysis of antigen presentation and T-cell recognition reveals restricted immune responses in melanoma. Cancer Discov 2018;8: 1366–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kassem A, Schöpflin A, Diaz C, Weyers W, Stickeler E, Werner M, et al. Frequent detection of Merkel cell polyomavirus in human Merkel cell carcinomas and identification of a unique deletion in the VP1 gene. Cancer Res 2008;68:5009–13. [DOI] [PubMed] [Google Scholar]
- Kaufman HL, Russell J, Hamid O, Bhatia S, Terheyden P, D’Angelo SP, et al. Avelumab in patients with chemotherapy-refractory metastatic Merkel cell carcinoma: a multicentre, single-group, open-label, phase 2 trial. Lancet Oncol 2016;17:1374–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kiepiela P, Leslie AJ, Honeyborne I, Ramduth D, Thobakgale C, Chetty S, et al. Dominant influence of HLA-B in mediating the potential co-evolution of HIV and HLA. Nature 2004;432:769–75. [DOI] [PubMed] [Google Scholar]
- Kikuchi S, Wada O, Nakajima T, Nishi T, Kobayashi O, Konishi T, et al. Serum anti-Helicobacter pylori antibody and gastric carcinoma among young adults. Research Group on Prevention of Gastric Carcinoma among Young Adults. Cancer 1995;75:2789–93. [DOI] [PubMed] [Google Scholar]
- 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. Clin Cancer Res 2019;25:5961–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koutsakos M, McWilliam HEG, Aktepe TE, Fritzlar S, Illing PT, Mifsud NA, et al. Downregulation of MHC class I expression by influenza A and B viruses. Front Immunol 2019;10:1158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koyama S, Akbay EA, Li YY, Herter-Sprie GS, Buczkowski KA, Richards WG, et al. Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat Commun 2016;7: 10501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kreiter S, Vormehr M, van de Roemer N, Diken M, Löwer M, Diekmann J, et al. Mutant MHC class II epitopes drive therapeutic immune responses to cancer. Nature 2015;520:692–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kurzen H, Kaul S, Egner U, Deichmann M, Hartschuh W. Expression of MUC 1 and Ep-CAM in Merkel cell carcinomas: implications for immunotherapy. Arch Dermatol Res 2003;295:146–54. [DOI] [PubMed] [Google Scholar]
- Lastwika KJ, Kargl J, Zhang Y, Zhu X, Lo E, Shelley D, et al. Tumor-derived autoantibodies identify malignant pulmonary nodules. Am J Respir Crit Care Med 2019;199:1257–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017;357:409–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med 2015;372:2509–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leach DR, Krummel MF, Allison JP. Enhancement of antitumor immunity by CTLA-4 blockade. Science 1996;271:1734–6. [DOI] [PubMed] [Google Scholar]
- 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 Immunol Res 2013;1:54–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Logan AC, Gao H, Wang C, Sahaf B, Jones CD, Marshall EL, et al. High-throughput VDJ sequencing for quantification of minimal residual disease in chronic lymphocytic leukemia and immune reconstitution assessment. Proc Natl Acad Sci USA 2011;108:21194–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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;7:1727–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marincola FM, Jaffee EM, Hicklin DJ, Ferrone S. Escape of human solid tumors from T-cell recognition: molecular mechanisms and functional significance. Adv Immunol 2000;74:181–273. [DOI] [PubMed] [Google Scholar]
- McMahan RH, Golden-Mason L, Nishimura MI, McMahon BJ, Kemper M, Allen TM, et al. Tim-3 expression on PD-1+ HCV-specific human CTLs is associated with viral persistence, and its blockade restores hepatocyte-directed in vitro cytotoxicity. J Clin Invest 2010;120:4546–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mehta A, Kim YJ, Robert L, Tsoi J, Comin-Anduix B, Berent-Maoz B, et al. Immunotherapy resistance by inflammation-induced dedifferentiation. Cancer Discov 2018;8:935–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miller NJ, Church CD, Fling SP, Kulikauskas R, Ramchurren N, Shinohara MM, et al. Merkel cell polyomavirus-specific immune responses in patients with Merkel cell carcinoma receiving anti-PD-1 therapy. J Immunother Cancer 2018;6:131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Naito Y, Saito K, Shiiba K, Ohuchi A, Saigenji K, Nagura H, et al. CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer. Cancer Res 1998;58:3491–4. [PubMed] [Google Scholar]
- Nghiem P, Bhatia S, Lipson EJ, Sharfman WH, Kudchadkar RR, Brohl AS, et al. Durable tumor regression and overall survival in patients with advanced Merkel cell carcinoma receiving pembrolizumab as first-line therapy. J Clin Oncol 2019;37:693–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nghiem PT, Bhatia S, Lipson EJ, Kudchadkar RR, Miller NJ, Annamalai L, et al. PD-1 blockade with pembrolizumab in advanced Merkel-cell carcinoma. N Engl J Med 2016;374:2542–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pai CS, Huang JT, Lu X, Simons DM, Park C, Chang A, et al. Clonal deletion of tumor-specific T cells by interferon-gamma confers therapeutic resistance to combination immune checkpoint blockade. Immunity 2019;50:477–92.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paulson KG, Carter JJ, Johnson LG, Cahill KW, Iyer JG, Schrama D, et al. Antibodies to Merkel cell polyomavirus T antigen oncoproteins reflect tumor burden in Merkel cell carcinoma patients. Cancer Res 2010;70:8388–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. Am J Clin Pathol 2014a;142:452–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. J Clin Oncol 2011;29:1539–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paulson KG, Lahman MC, Chapuis AG, Brownell I. Immunotherapy for skin cancer. Int Immunol 2019;31:465–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paulson KG, Lemos BD, Feng B, Jaimes N, Peñas PF, Bi X, et al. Array-CGH reveals recurrent genomic changes in Merkel cell carcinoma including amplification of L-myc. J Invest Dermatol 2009;129:1547–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paulson KG, Lewis CW, Redman MW, Simonson WT, Lisberg A, Ritter D, et al. Viral oncoprotein antibodies as a marker for recurrence of Merkel cell carcinoma: a prospective validation study. Cancer 2017;123:1464–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paulson KG, Perdicchio M, Kulikauskas R, Wagener F, Church C, Bui K-T, et al. Augmentation of adoptive T-cell therapy for Merkel cell carcinoma with avelumab. J Clin Oncol 2017;35(Suppl. 15). 3044–3044. [Google Scholar]
- Paulson KG, Tegeder A, Willmes C, Iyer JG, Afanasiev OK, Schrama D, et al. Downregulation of MHC-I expression is prevalent but reversible in Merkel cell carcinoma. Cancer Immunol Res 2014b;2:1071–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paulson KG, Voillet V, McAfee MS, Hunter DS, Wagener FD, Perdicchio M, et al. Acquired cancer resistance to combination immunotherapy from transcriptional loss of class I HLA. Nat Commun 2018;9:3868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Penn I, First MR. Merkel’s cell carcinoma in organ recipients: report of 41 cases. Transplantation 1999;68:1717–21. [DOI] [PubMed] [Google Scholar]
- Phares TW, Stohlman SA, Hinton DR, Bergmann CC. Enhanced CD8 T-cell anti-viral function and clinical disease in B7-H1-deficient mice requires CD4 T cells during encephalomyelitis. J Neuroinflammation 2012;9:269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pos Z, Spivey TL, Liu H, Sommariva M, Chen J, Wunderlich JR, et al. Longitudinal study of recurrent metastatic melanoma cell lines underscores the individuality of cancer biology. J Invest Dermatol 2014;134:1389–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ritter C, Fan K, Paulson KG, Nghiem P, Schrama D, Becker JC. Reversal of epigenetic silencing of MHC class I chain-related protein A and B improves immune recognition of Merkel cell carcinoma. Sci Rep 2016;6:21678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015;348:124–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Robins H Immunosequencing: applications of immune repertoire deep sequencing. Curr Opin Immunol 2013;25:646–52. [DOI] [PubMed] [Google Scholar]
- Rock KL, Reits E, Neefjes J. Present yourself! By MHC class I and MHC class II molecules. Trends Immunol 2016;37:724–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seiwert TY, Zuo Z, Keck MK, Khattri A, Pedamallu CS, Stricker T, et al. Integrative and comparative genomic analysis of HPV-positive and HPV-negative head and neck squamous cell carcinomas. Clin Cancer Res 2015;21:632–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shahzad N, Shuda M, Gheit T, Kwun HJ, Cornet I, Saidj D, et al. The Tantigen locus of Merkel cell polyomavirus downregulates human toll-like receptor 9 expression. J Virol 2013;87:13009–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shankaran V, Ikeda H, Bruce AT, White JM, Swanson PE, Old LJ, et al. IFN-gamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature 2001;410:1107–11. [DOI] [PubMed] [Google Scholar]
- Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 2017;168:707–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shimizu S, Hiratsuka H, Koike K, Tsuchihashi K, Sonoda T, Ogi K, et al. Tumor-infiltrating CD8+ T-cell density is an independent prognostic marker for oral squamous cell carcinoma. Cancer Med 2019;8:80–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. Clin Cancer Res 2012;18:2872–1. [DOI] [PubMed] [Google Scholar]
- Spradling PR, Xing J, Williams R, Masunu-Faleafaga Y, Dulski T, Mahamud A, et al. Immunity to hepatitis B virus (HBV) infection two decades after implementation of universal infant HBV vaccination: association of detectable residual antibodies and response to a single HBV challenge dose. Clin Vaccine Immunol 2013;20:559e–1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spurgeon ME, Lambert PF. Merkel cell polyomavirus: a newly discovered human virus with oncogenic potential. Virology 2013;435:118–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sucker A, Zhao F, Real B, Heeke C, Bielefeld N, Maben S, et al. Genetic evolution of T-cell resistance in the course of melanoma progression. Clin Cancer Res 2014;20:6593–604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tamborero D, Rubio-Perez C, Muiños F, Sabarinathan R, Piulats JM, Muntasell A, et al. A pan-cancer landscape of interactions between solid tumors and infiltrating immune cell populations. Clin Cancer Res 2018;24: 3717–28. [DOI] [PubMed] [Google Scholar]
- Thorsson V, Gibbs DL, Brown SD, Wolf D, Bortone DS, Ou Yang TH, et al. The immune landscape of cancer. Immunity 2018;48:812–30.e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walboomers JM, Jacobs MV, Manos MM, Bosch FX, Kummer JA, Shah KV, et al. Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. J Pathol 1999;189:12–9. [DOI] [PubMed] [Google Scholar]
- 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 Res 2015;75:5228–34. [DOI] [PubMed] [Google Scholar]
- Wu D, Sherwood A, Fromm JR, Winter SS, Dunsmore KP, Loh ML, et al. High-throughput sequencing detects minimal residual disease in acute T lymphoblastic leukemia. Sci Transl Med 2012;4:134ra63. [DOI] [PubMed] [Google Scholar]
- Yadav M, Jhunjhunwala S, Phung QT, Lupardus P, Tanguay J, Bumbaca S, et al. Predicting immunogenic tumour mutations by combining mass spectrometry and exome sequencing. Nature 2014;515:572–6. [DOI] [PubMed] [Google Scholar]
- Yarchoan M, Albacker LA, Hopkins AC, Montesion M, Murugesan K, Vithayathil TT, et al. PD-L1 expression and tumor mutational burden are independent biomarkers in most cancers. JCI Insight 2019;4:e126908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med 2017;377:2500e1. [DOI] [PMC free article] [PubMed] [Google Scholar]