Next Article in Journal
Towards Novel Non-Invasive Colorectal Cancer Screening Methods: A Comprehensive Review
Next Article in Special Issue
Cannabinoid Receptor Type-2 in B Cells Is Associated with Tumor Immunity in Melanoma
Previous Article in Journal
The Leloir Cycle in Glioblastoma: Galactose Scavenging and Metabolic Remodeling
Previous Article in Special Issue
Cutaneous Squamous Cell Carcinoma in the Age of Immunotherapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Find the Flame: Predictive Biomarkers for Immunotherapy in Melanoma

1
CRO Aviano National Cancer Institute IRCCS, 33081 Aviano, Italy
2
DSM—Department of Medical Sciences, University of Trieste, 34123 Trieste, Italy
3
Department of Medicine (DAME), University of Udine, 33100 Udine, Italy
4
Dipartimento di Oncologia, Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy
5
Department of Dermatology, University of Trieste, 34123 Trieste, Italy
*
Author to whom correspondence should be addressed.
Cancers 2021, 13(8), 1819; https://doi.org/10.3390/cancers13081819
Submission received: 8 March 2021 / Revised: 24 March 2021 / Accepted: 30 March 2021 / Published: 10 April 2021
(This article belongs to the Special Issue Molecular Mechanisms of Skin Cancer)

Abstract

:

Simple Summary

Immunotherapy has revolutionized the therapeutic landscape of melanoma. However, the absence of clinically validated predictive biomarkers is one of the major causes of unpredictable efficacy of immunotherapy. Indeed, the availability of predictive biomarkers could allow a better stratification of patients, suggesting which type of drugs should be used in a certain clinical context and guiding clinicians in escalating or de-escalating therapy. However, the difficulty in obtaining clinically useful predictive biomarkers reflects the deep complexity of tumor biology. Herein, we review the available literature to depict the most useful or promising biological biomarker able to predict immunotherapy response in melanoma. We also make a meta-analysis regarding PDL1 expression in melanoma and immune checkpoint response.

Abstract

Immunotherapy has revolutionized the therapeutic landscape of melanoma. In particular, checkpoint inhibition has shown to increase long-term outcome, and, in some cases, it can be virtually curative. However, the absence of clinically validated predictive biomarkers is one of the major causes of unpredictable efficacy of immunotherapy. Indeed, the availability of predictive biomarkers could allow a better stratification of patients, suggesting which type of drugs should be used in a certain clinical context and guiding clinicians in escalating or de-escalating therapy. However, the difficulty in obtaining clinically useful predictive biomarkers reflects the deep complexity of tumor biology. Biomarkers can be classified as tumor-intrinsic biomarkers, microenvironment biomarkers, and systemic biomarkers. Herein we review the available literature to classify and describe predictive biomarkers for checkpoint inhibition in melanoma with the aim of helping clinicians in the decision-making process. We also performed a meta-analysis on the predictive value of PDL-1.

1. Introduction

Historically, melanoma was described as a highly lethal disease [1]. However, in the last 10 years, several new therapeutic strategies have been shown to increase survival in the metastatic and adjuvant settings [2,3,4]. Among the contemporary available treatments, immunotherapy (e.g., checkpoint inhibitors (ICIs)) has demonstrated an impressive efficacy. Indeed, checkpoint inhibition, which was described as “breakthrough of the year 2013” [5], has also shown to be able to control melanoma in a long-term fashion [4]. These long-term remissions have opened up the possibility of a virtual curative effect of immunotherapy. However, to date, reliable biomarkers able to predict checkpoint inhibition efficacy in melanoma are lacking.
The availability of predictive biomarkers for immunotherapy could help clinicians choose the best therapeutic strategy for melanoma patients. Practically, where a benefit from immunotherapy is predicted, treatment de-escalation might be possible. On the contrary, melanomas predicted to be refractory to checkpoint inhibition could benefit from alternative treatments (i.e., targeted therapies) or a treatment escalation. Moreover, in some patients treated with immunotherapy, a precocious progression of the disease has been described. Those patients who are called hyper-progressors seem to have a poor prognosis [6]. Predictive biomarkers could help to identify these patients and treat them accordingly.
Predictive biomarkers for immunotherapy can be classified into three major groups: tumor-intrinsic biomarkers, which are expressed by tumor cells (e.g., PD-1/PDL-1, tumor mutational burden); tumor microenvironment biomarkers (e.g., tumor-infiltrating lymphocyte); and systemic biomarkers (e.g., circulating factors, microbiota).
Herein we review the available literature to classify and describe predictive biomarkers for checkpoint inhibition in melanoma to help clinicians in the decision-making process (Table 1). In particular, we focus on neoadjuvant and metastatic settings, where an evaluation of response rate can be performed. Moreover, we consider trials with anti-PD-1/PDL-1 and/or anti-CTLA-4.

2. Materials and Methods

2.1. Searching Details

For the meta-analysis we searched the database PubMed using the following keywords: (1) PD-L1 expression melanoma immunotherapy response; (2) melanoma keynote; (3) PD-L1 immunohistochemistry melanoma predictive; (4) PD-L1 nivolumab response melanoma; (5) PD-L1 expression predictive immunotherapy melanoma. We searched articles published up to November 2020. The reference lists were also carefully checked to identify additional eligible studies. All analyses were carried out with previously published data; consequently, no ethical approval or patient consent was necessary in this study. For the other manuscript sections, we performed scoping research.

2.2. Selection Criteria

Studies were included if they met the following criteria: (1) inclusion of patients diagnosed with confirmed melanoma; (2) treatment with immune checkpoint inhibitors; (3) detection of PD-L1 expression in the melanoma tissue, including metastases, by immunohistochemistry (IHC) irrespective of the antibody clone used; (4) identification of a definite cutoff value in the analysis of PD-L1 expression; (5) availability of RECIST 1.1 data (responders = complete + partial response according to RECIST for solid tumors) in PD-L1-positive and in PD-L1-negative patients; (5) publications in English language. Determination of PD-L1 expression in melanoma was based on most analyzed studies by the percentage of PD-L1 expressing tumor cells; however, in a minority of studies, inflammatory cells in nest of tumor cells were also counted.
The exclusion criteria were as follows: (1) case reports, reviews (systematic reviews were retained), letters, and correspondences; (2) studies without available or usable information; (3) studies including adjuvant therapy with immune checkpoint inhibitors; (4) studies lacking PD-L1 or RECIST data; (5) animal studies; (6) publications in languages other than English; (7) duplicate studies.

2.3. Data Extraction

One researcher (S.B.) extracted basic information from the included studies, also analyzing supplementary data where available. The following data were extracted from eligible studies: the first author’s name, publication year, sample size, detection method, antibody used (where available), treatment, cutoff values, and number of responders and non-responders in the group of PD-L1-positive and PD-L1-negative patients.

2.4. Statistical Analysis

The percent of responders and non-responders in the group of PD-L1-positive and PD-L1-negative was used to set up the meta-analysis for binary data. The association between PD-L1 expression and response to ICI therapy was assessed by odds ratios (ORs) and their 95% CIs. The I2 metric was used to inspect the statistical heterogeneity of the data. A p value of less than 0.1 or an I2 value of more than 50% indicated significant heterogeneity, and a random effects model was employed for calculation. All statistical analyses were carried out using Stata version 16.0 (STATA Corp., College Station, TX, USA). p < 0.05 was considered to indicate statistical significance.

2.5. Results

In PubMed, after removal of duplicates, 309 publications were retained. From these, 52 articles were read as full text. Of these, 22 articles were retained as they fulfilled the inclusion criteria.

3. Tumor-Intrinsic Biomarkers

3.1. Tumor Mutational Burden

Tumor mutational burden (TMB) is an index that summarizes the mutational load of a tumor [84]. Since a high number of mutations could translate into a high number of neoantigens that the immune system can recognize, it has been hypothesized that TMB could act as a proxy for ICI effectiveness.
The definition and the methods for the detection of TMB are evolving [85]. Initially, the tumor mutational load was searched in the tumor specimen through the costly whole-exome sequencing (WES) technique, which takes into account the non-synonymous mutations in the coding regions of the genome [86]. More recently, to reduce costs and increase the breadth of investigation, next-generation sequencing (NGS) has been used on restricted gene panels in place of WES [10,14,87]. Among NGS methods, two tests have recently received approval from the FDA: FoundationOne CDx [88] (Cambridge, USA) and MSK-IMPACT [89] (Memorial Sloan Kettering, USA). The first one profiles 324 genes (corresponding to 1.1 Mb of coding genome) and also includes short indel in intronic regions, which are excluded from whole-exome sequencing. MSK-IMPACT analyzes 468 cancer-related genes for exonic mutations (approximately 1.2 Mb). More recently, in an attempt to simplify the search for TMB, a liquid biopsy approach has been used. Although at the beginning a discordance between tissue- and blood-based approaches seemed evident [90], a convergence of results might have been reached [91,92,93]. The detection of TMB in liquid biopsy could offer several advantages over the tissue-based approach. For example, it is a non-invasive procedure that allows serial sampling over time and the detection of TMB status in patients who do not have adequate tissue available.
Independently from the method used to analyze TMB, a linear correlation between TMB levels and the efficacy of ICIs seems to exist [94]. However, there is still a lack of consensus regarding the cutoff for the definition of high (TMB-H), intermediate(TMB-I), and low levels (TMB-L) of TMB. For example, FoundationOne CDx uses the threshold of 20 mutations/Mb for the definition of TMB-H, 6–19 mutations/Mb for the TMB-I, and ≤5 mutations/Mb for TMB-L [88,95]. Nonetheless, the FDA approved the agnostic use of pembrolizumab for tumors with a TMB threshold of at least 10 mutations/Mb. Moreover, since the mutational load varies considerably between histologies, the use of the top 20% value of TMB for each histology has been proposed [89]. However, a harmonization between methodologies is highly needed and is ongoing [94,96].
The observation that TMB levels show a linear correlation with the response to ICIs seems quite logical since a higher number of mutations could translate into a higher number of neoantigens that the immune system could recognize. However, this syllogism is somehow imperfect, as even tumors with a TMB-H have a response rate (RR) to ICIs of only 45% [97]. A possible explanation might reside in the variable presentation of neoantigens to lymphocytes that is strictly correlated to the proper functioning of MHC machinery (see below).
In melanoma, coherently with other cancer types, TMB levels correlate to ICI RR both with anti-PD-1/PDL-1 and anti-CTLA-4 therapies. As shown by Cristescu et al. [13], melanomas with TMB-H have a RR to pembrolizumab of 42% versus only 9% of melanomas with TMB-L. However, in that study, a T-cell inflamed gene-expression profile emerged as another variable influencing RR to pembrolizumab, suggesting that TMB might be used in conjunction with other biomarkers to predict ICI response better. Similar results have been shown by Van Allen et al. for ipilimumab [7].
Even though TMB-H alone can identify a subset of tumors particularly sensitive to ICIs, it should be highlighted that TMB-L cancers could also derive benefits from immunotherapies, albeit in a small percentage of patients (5%) [97]. Theoretically, even in a context of low mutational burden, strong immunogenic neoantigens could be generated, albeit with a lower probability compared to a TMB-H context.

3.2. MHC-I and MHC-II

Major histocompatibility complex (MHC) is the cellular machinery that is exposed on the cellular surface of small peptides that can be recognized by immune cells [98,99]. These, in turn, could become activated if the peptides that they recognize are identified as “non-self”. MHC can be classified as type I and type II [98,99]. The first can mediate the activation of CD8 lymphocytes subpopulation, while MHC class II regulates the activation of CD4 lymphocytes.
As stated above, the roles of TMB and MHC are quite intertwined. In order to be recognized by the immune system, a neoantigen has to be exposed on a cancer cell surface through the MHC machinery. While a high TMB increases the chance to generate immunogenic neoantigens, a functioning MHC machinery increases the chance that these immunogenic neoantigens will be identified by immune cells. The type of immune response after the recognition of the neoantigen by the immune cells may vary. CD8 lymphocytes, for instance, can exert a direct cytotoxic activity against cancer cells, while CD4 lymphocytes can stimulate a broad inflammatory response through the production of interferon-gamma [98,99].
In melanoma, several pieces of evidence show a predictive role for MHC to immunotherapy. However, a different predictive role between MHC class I and II seems to exist. In the work of Johnson et al., MHC class II positive-melanomas have a better response rate, progression-free survival, and overall survival in anti-PD-1 therapy [18,19]. However, as shown by Liu et al., the predictive role of MHC class II expression in melanoma cells in anti-PD-1 might be restricted in patients with previous exposure to an anti-CTLA-4 [17]. Interestingly, Rodig et al. showed a differential predictive role in immunotherapy for MHC class I and II [20]. While class II seems to predict benefit from anti-PD-1 therapy, MHC class I could be predictive for anti-CTLA-4 drugs. Although checkpoint inhibitors’ anticancer effect has historically been linked to the activation of cytotoxic CD8 lymphocytes [100], mounting evidence highlights a pivotal role for CD4, especially during an anti-PD-1 therapy [101]. Finally, the loss of B2M, a crucial protein belonging to the MHC class I system, has been described as a resistance mechanism to both anti-CTLA-4 and anti-PD-1 drugs [21].
Of note, there is still a lack of consensus regarding the cutoff for the definition of high- or low-MHC expressing tumors. Moreover, several proteins belonging to MHC machinery could be evaluated, and this could jeopardize the identification of MHC-positive or -negative cancers [98,99].

3.3. PD-1/PDL-1

Programmed cell death 1 receptor (PD-1) is a checkpoint molecule present on T-cells, B-cells, and natural killer cells (NKs), which can interact with its ligands: PD-L1, expressed on tumor cells, and PD-L2, which is mainly present on hematopoietic cells [102]. The binding of PD-1 with its ligands is responsible for T-cell dysfunction and/or neutralization. Thus, overexpression or amplification of PD-L1 by tumor cells possibly diminishes immune antitumor response [102] with a mechanism of adaptive resistance [103]. Therefore, theoretically, patients whose tumors have a high level of PD-L1 should benefit better from ICI therapy, especially for those drugs targeting the PD-1/PD-L1 axis. PD-L1 is, indeed, the most commonly recognized biomarker to predict immunotherapy response in patients with different solid tumors, including cutaneous melanoma [104]. Immunohistochemical tests to assess PD-L1 expression are approved by the US Food and Drug Administration (FDA) as companion diagnostic assays for immunotherapy response for several solid tumors, but not for melanoma [105,106]. Staining issues related to specific antibody clones (28-8 vs. 22C3) as well as staining protocols have been successfully fixed, and PD-L1 staining has been highly harmonized and validated [107]. Nonetheless, immunohistochemistry using clone SP142 returned the lowest positivity level in most solid tumors [108]. In melanoma, positivity cutoffs for PD-L1 are 1% (corresponding to Melscore 2) [109] or 5% of tumor cells [110]. Alternatively, in addition to tumor cells, inflammatory cells in nests of tumor cells have been sometimes included in the count [111]. Rarely, melanin pigment in primary or metastatic melanomas can impact PD-L1 immunohistochemistry interpretation [108]. The meta-analysis carried out in the present review was set using as the endpoint of RECIST response rate (complete or partial response) to ICI therapy with respect to the PD-L1 expression by immunohistochemistry irrespective of the antibody clone and the cutoff (details of the inclusion criteria are reported in the Supplementary Material). Using selection criteria, 23 studies, including results for 4710 patients, were retained for the analysis [16,19,43,44,45,46,47,70,75,109,110,111,112,113,114,115,116,117,118,119,120,121]. Despite the heterogeneity in the treatments and staining cutoffs, our analysis shows that patients with PD-L1-positive melanomas/recurrences have, on average, a higher rate of response to ICI, as shown in Figure 1 and Table 2. By sorting data per ICI treatment, no significant difference in the overall response rate between PD-L1-positive and -negative melanomas was observed for ipilimumab treated patients, in line with the fact that it is an anti-CTLA-4 molecule. However, for pembrolizumab, nivolumab, and combined regimens, there is a significantly higher benefit in patients with positive PD-L1 melanomas. Notwithstanding, PD-1/PD-L1 inhibitors appear to also have activity in subsets of patients who do not meet IHC positivity to PD-L1 [122]. This is mainly because the PD-L1/PD-1 axis is not the only player in response to checkpoint inhibition [103]. It is manifest that PD-L1 status alone cannot be used as a reliable biomarker of therapeutic response to anti-PD-1 immunotherapy in melanoma. PD-L1 expression is dynamic and transient with possible intra-patient and intratumor heterogeneity [123], and it can be affected by many factors, including previous therapies and the presence of tumor-infiltrating immune cells [109]. Therefore, in melanoma, PD-L1 status should be combined with other criteria, such as TMB [104], CD8, and PD-1 in T-cells [124], to better describe the response to PD-1/PD-L1 inhibitors.

4. Microenvironment Biomarkers

4.1. TILs

Tumor-infiltrating lymphocytes (TILs) are a heterogeneous population comprising several cell subtypes such as effector (CD8+) and regulatory (CD4+ and CD25/FoxP3+) T-lymphocytes, natural killer cells (NKs), dendritic cells, and macrophages [124]. They were described by Clark et al. [126] in 1969, and their presence in tumoral specimens was subsequently found to be positively associated with better prognosis during the vertical growth phase of high-risk melanoma [127,128,129].
Beyond the dichotomic evaluation of their presence in the tumoral specimen, the following studies have focused on their distribution, phenotype, and state of activation, assuming that these characteristics should reflect the disease’s host reaction and therefore could have an impact on the clinical outcome [130,131,132]. Type, density, and location of immune cells are the variables included in the Immunoscore [133], the predictive role of which was investigated in the MISIPI trial [33]. Surprisingly, CD3+, CD8+, CD20+, CD163+, and FoxP3+ T-cells, both intratumoral (CT) and peritumoral (IM), had no impact on response to treatment in melanoma patients (pts) [33,34,134], underlying the need for functional status analysis of immune cells.
In one of the first biomarker-discovery trials, Hamid et al. [27], enrolled 82 pts with unresectable melanoma (stage IV or III) treated with ipilimumab. Tumor biopsies were performed before the start of treatment and after the second drug administration. Afterwards, pts were divided into a benefit (complete/partial response and stable disease) and non-benefit groups, based on radiological response. In the benefit group, 57.1% of patients had a post-treatment increase in TILs compared to baseline (OR 13.2), and FOXP3+ positivity was detected in 75% of pretreatment samples (vs. 36% in the non-benefit group). Moreover, a high basal indoleamine-2,3-dioxygenase (IDO) expression was related to a favorable outcome. Despite the historically immunosuppressive role of IDO, considerable evidence highlighted that its expression is induced by IFN-γ signaling. From this perspective, a tumoral microenvironment with high IDO expression could reflect an inflamed phenotype [135,136].
In another study, Daud et al. [28] isolated samples of metastatic melanoma before anti-PD-1 therapy and assessed the presence of effector (FOXP3-) and regulatory (FOXP3+) CD4+ T-cells, CD8+ cytotoxic lymphocytes (CTLs), and the expression of PD-1, PD-L1, and CTLA-4 on each cell group. A presence of at least 20% of CTLA-4hi PD-1hi CTLs was related to a greater response rate (RR) (85.7% vs. 0%) and PFS (31.6 months vs. 9.6 months, p = 0.017) compared to low CTLA-4hi PD-1hi CTLs. Intriguingly, CTLA-4hi PD-1hi CTLs revealed a partially exhausted phenotype with a maintained production of INF-γ but a reduction of IL-2 and TNF-α. Other T-cell subsets showed no impact on RR.
Consistently, Tumeh et al. [29], in a trial including 46 pts with advanced melanoma treated with pembrolizumab, reported an increase in intratumoral CD8+ cells in pts responsive to the treatment, according to the radiological regression. Regarding the distribution of TILs, CD8+ lymphocytes were more represented at the invasive margin (IM) in pretreatment samples than the non-responder group, suggesting their pivotal role in tumor regression. In the responder group, pembrolizumab also determined an increase of Ki-67-positive CD8+ cells in the tumor center (TC). Conversely, CD4+ cell density, both in TC and IM, showed a low impact on the prediction of treatment response, as previously reported. These data were successively confirmed in the neoadjuvant setting with growing interest in exhausted T-cells and the relation between TILs and circulating lymphocytes [30].
The role of CD4+ lymphocytes might be more important than initially assumed [101]. Indeed, mounting evidence suggests that CD4 lymphocytes could exert a direct and indirect anticancer effect, such as after a vaccination with cancer peptides [101]. Moreover, a trial testing anti-PD-1 drugs in an advanced setting reported a statistically significant association between intratumoral CD4+ cell absolute count and RR [32]. This discordance may be due to the plasticity of CD4+ T lymphocytes, capable of switching from a regulatory to an effector phenotype [132,137]. From this perspective, the ratio between the T-cell populations could provide a more detailed overview of immune balance [132,138]. Of note, CD4 lymphocytes are a heterogeneous class of T-cells [101]. More research is needed to identify the antitumoral effect of each subpopulation.
Natural killer cells (NKs) are usually rare in melanoma tissues, and their predictive and prognostic roles are still debatable [132,139]. Physiologically, NKs are activated by several mediators such as adhesion molecules, soluble extracellular factors, proinflammatory cytokines, and danger-associated molecular patterns (DAMPs) and exert their effector function through the apoptosis of those cells downregulating MHC I molecules, cancer cells included [78,92,132,139].
Preclinical evidence have been emerging, and a high number of NKs seems to predict the response to IT through the production of the FMS-like tyrosine kinase-3 ligand (FLT3L). This hematopoietic cytokine acts as a growth factor [140], which stimulates the intratumoral dendritic cells (DCs) [141]. On the other hand, a small trial found a positive relationship between low circulating total NKs number and ipilimumab response [69]. Intratumoral NKs showed several transcriptional profiles, probably influenced and dynamically regulated by the tumoral microenvironment. Additional studies are required to clarify which subtype may be related to the response to IT and its relationship with NK tumoral levels [142].
Type-2 tumor-associated macrophages (TAMs) are a substantial component of TILs and exert an immune-suppressive function limiting the efficacy of IT through the inhibition of CTLs. Currently, no studies have been conducted in vivo, but preclinical evidence showed promising results with TAM reprogramming strategies [143,144].

4.2. Gene Signatures

The host response to the tumor consists of a continuum of innate and adaptive immune reactions, where the immunosurveillance and the immune escape phenomena interplay [145]. Therefore, the quantification and the descriptive analysis of the immune contexture [146] provide a partial overview of the treatment response’s complex mechanism. Those morphological features have been integrated with the underlined genomic landscape analysis, and the role of immune-related genes has been broadly investigated. As a result, the concept of gene-expression profile (GEP) has emerged [147].
INF-γ signature certainly retains the most robust evidence, and several studies have proved its role in establishing an inflamed tumoral microenvironment, both in a palliative and neoadjuvant setting [30,35,148,149].
Ayers et al. [36] conducted a preliminary analysis on 19 pts enrolled in the KEYNOTE-001 trial, building up a 10-gene IFN signature (IFNG, STAT1, CCR5, CXCL9, CXCL10, CXCL11, IDO1, PRF1, GZMA, and MHCII HLA-DRA) which allowed responders to be discriminated from non-responders. This signature was expanded to 28 genes, including some related to antigen presentation, chemokine and cytokine activity, and immunomodulatory factors, and was later validated in a cohort of 62 pts. Both these signatures positively correlated to RR and PFS and confirmed their predictive value to anti-PD-1 therapy. Consistently with the studies reported above, in the OpacinNeo study, a low IFN-γ expression seems to be related to a higher risk of relapse [148].
Besides IFN-γ signature, T-cell-activating genes are involved in response to IT as well.
A pretreatment GEP with high expression of genes related to T-cell infiltration seems to enhance the immune response during the treatment, underlying the importance of a pre-existing inflamed phenotype [150].
In addition to the GEP basal evaluation, the post-treatment genomic modifications have also been evaluated. Both pembrolizumab [30] and ipilimumab [27] were associated with a post-treatment expression of immune-response related genes (such as those coding for immunoglobulins, granzyme B, perforin-1, granulysin, CD8 b-subunit, and T-cell receptor-a and -b subunits) with an impact on recurrence-free survival.
Chen et al. [37] analyzed 46 pts treated with anti-PD-1 agents, progressed to the previous anti-CTLA-4 drug, and investigated their immune profile with an NGS panel. Early on-treatment samples of responders showed 411 significantly differentially expressed genes, predominantly upregulated compared to non-responders, including those coding for chemokines, INF-γ pathway mediators, and adhesion molecules. Moreover, a longitudinal analysis was conducted, and responders to the anti-PD-1 drug revealed an upregulation of 376 genes involved in antigen presentation, T-cell activation, and T-cell homing.
Similarly, Gide et al. [38] performed transcriptomic and immune profiling on 158 melanoma samples and identified two gene clusters associated with better outcomes (PFS) with anti-PD-1 monotherapy. In particular, IFN-related genes (such as TBX21, STAT1, IRF1, TNF, and IFNG) and tumor-infiltrating T-cell genes (CCL5, CXCL13, and IL-2) were highly expressed, suggesting activated T-cells enriched tumoral microenvironment in responders with IFN-secretive phenotype.
Focusing on immune-escape rather than immune-(re)activation, Jiang et al. [151] identified a signature (TIDE) to measure T-cell dysfunction and T-cell exclusion and showed that it could discriminate between responders and non-responders especially in those with high CTL tumoral infiltration.
In conclusion, the emerging evidence on GEP and gene signatures highlights the concept of a genomic (constitutive) predisposition, closely tied to the immune cell infiltration in the tumoral microenvironment. From this perspective, these aspects should be evaluated in combination to provide an exhaustive overview of the complex phenomenon of response to ICIs.
However, predictive gene signatures have not been consistent between different studies, suggesting that there might not exist a unique molecular pattern that predicts response to immunotherapy. From this perspective, an inclusive panel of gene signatures might be able to better predict the outcome from checkpoint inhibitors.

5. Systemic Biomarkers

5.1. Circulating Factors

Elevated serum lactate dehydrogenase (LDH) is an established surrogate of high tumor burden and an independent adverse prognostic factor in stage IV malignant melanoma patients [152,153]. LDH is included in the eighth edition of the AJCC (American Joint Committee on Cancer) for the malignant melanoma staging system [154]. Several studies proved that patients with an elevated baseline LDH had a significantly shorter overall survival (OS) than patients with normal LDH during ICI therapy [39,40,152]. Moreover, the relative changes in LDH levels during the first ICI treatment weeks are early markers for response and OS [39,40,41]. Preclinical data suggest that LDH, through its interaction in glycolysis and hypoxia mechanisms, contributes to an immune-suppressive microenvironment [155,156]. Even the calcium-binding protein S100B has been used in staging melanoma, establishing prognosis, evaluating treatment success, and predicting relapse [157,158]. High values of S100B and LDH at baseline are associated with the poor outcome either during pembrolizumab alone or with ICI combined therapy [40]. C-reactive protein (CRP), a well-known inflammation index, is another independent prognostic marker in patients with melanoma, associated with shorter OS and melanoma-specific survival [159] and high tumor burden [160]. A recent analysis of CheckMate 064, 066, and 067 trials [42,161] highlighted that higher CRP baseline levels were associated with a weaker response and shorter survival after nivolumab alone, ipilimumab alone, or their combination. A trend towards decreased CRP levels during ipilimumab treatment and response and survival has also been reported [41,160]. Recently, anti-PD-1-treated patients with low serum concentration of hepatocyte growth factor (HGF) showed longer OS and PFS than those with high levels [162]. However, further evidence is needed to determine whether pharmacodynamic changes of these markers can be predictive of treatment efficacy or merely prognostic for survival [40,41,153].
Various cytokines in peripheral blood have been associated with ICI response. Pro-inflammatory IL-6 has been the most studied. High baseline IL-6 levels were associated with a poor response and shorter survival in patients receiving single-agent ICIs [42,48,161]. Moreover, in CheckMate-064, a decrease in IL-6 at 13 weeks was associated with longer OS than those with increasing levels [42]. A drop in IL-8 levels predicted response to anti-PD-1 [49], and baseline serum CXCL5 chemokine levels correlated with objective response to nivolumab in patients with malignant melanoma [50,51]. Furthermore, IL-8 and IL-6 play a role in myeloid-derived suppressor cell (MDSC) recruitment and expansion, further confirming their negative correlation with melanoma patient outcomes [163].
Higher soluble PDL-1 (sPDL-1) levels and sPDL-1 elevation predicted a higher likelihood of clinical benefit after 3 months of pembrolizumab, but they were associated with progressive disease during ipilimumab [52,53]. Recent studies indicated that PD-L1 expression in extracellular vesicles (exosomes), released from both T-cells and dendritic cells isolated from peripheral blood samples, reflects tumor characteristics [164]. Furthermore, PD-L1 is much easier to detect and quantify in circulating exosomes rather than in biopsies or when soluble in plasma [55]. Baseline increased PD-L1 and CD28 exosomal protein expression was associated with improved clinical response, PFS, and OS in ipilimumab-treated MM patients [54]. Moreover, a high increase in exosomal PD-L1 expression could predict tumor progression to ICI therapy [55].
Finally, soluble CTLA-4 levels were reported to be a positive predictive factor for ipilimumab therapy and to correlate with the best overall response [56].
Although promising, these blood-based biomarkers require large-scale prospective validation and cutoff definition.

5.2. Circulating Lymphocytes

Some insights in predicting survival and response during ICI therapy come from peripheral T-cell phenotype assessment. Higher absolute or relative lymphocyte, eosinophil, and neutrophil count at baseline and their increase during ICI treatment were associated with disease control and survival [41,57,58,59,60,165]. Higher baseline peripheral blood neutrophil to lymphocyte ratio (NLR), which may reflect chronic systemic inflammation, was strongly and independently associated with decreased PFS and OS with ICIs in several studies [42,61,62,63,64,65,66,67]. Alongside the simple absolute value and lymphocyte ratios, various T-cell phenotypes were investigated.
High frequencies of baseline CD8 effector-memory type 1 T-cells and normal baseline CD45RO+CD8+ T-cells levels correlate with longer OS (and only the first also higher clinical response rates) during ipilimumab [68,69].
Regulatory T-cells (Tregs), formerly known as suppressor T-cells, represent direct target cells of ipilimumab due to their constitutive CTLA-4 expression. Accordingly, baseline high FoxP3-Treg frequencies and their drop during ipilimumab were significantly associated with better survival [41,59]. An early increase in peripheral-blood Tregs and decreased antigen-specific T-cells have been associated with progression during nivolumab plus vaccine therapy [70].
Moreover, CD37 expression on circulating CD8+PD-1+ lymphocytes, involved in immune-suppressive adenosine production, seems to be a promising marker of clinical response to nivolumab [166]. Likewise, a lower than the median frequency of peripheral blood PD-1+CD56+ T-cells before starting anti-PD-1 is related to superior clinical response, longer PFS, and OS of stage IV melanoma patients [71].
Finally, an increase in relative numbers of CD4 and CD8 memory T-cell subsets and functionally active natural killer subsets were described as potential biomarkers for response to anti-CTLA-4 and anti-PD-1, respectively [72,165].
As it is known, prolonged and repeated antigen stimulation causes the T-cell exhaustion, consisting of loss of effector functions (such as IFNgamma production) and expression of multiple surface inhibitory receptors such as PD-1 and CTLA-4 [167,168]. Although it has been proposed as a promising biomarker when studied in TILs [168], peripheral blood expression of T-cell exhaustion markers does not distinguish between responders and non-responders to anti-PD-1 [73].
Finally, it is known that through the MHC-antigen complex-TCR interaction, a naïve T-cell is activated and clonally expanded. Therefore, the repertoire of T-cell receptors (TCRs) reflects the sum of previous exposures of unique antigens to the host. Intratumoral expanded TCR clonality at baseline and on-treatment was associated with response to ICIs [15,29]. Similarly, CTLA-4 and PD-1 blockade increased peripheral blood mononuclear cell TCR diversity as reflected in the higher number of unique TCR clonotypes in responders vs. non-responders [70,74,169]. However, conflicting results have been reported [11], and TCR analysis data seem more robust in tumoral specimens.

5.3. Microbiota

The human gastrointestinal tract harbors a number exceeding 1014 of symbiotic microorganisms, the gut microbiota, which exerts a marked influence on the host during homeostasis and disease [170]. There is growing evidence that gut microbiota composition and, in particular, specific bacterial species influence the efficacy and toxicity of ICI therapy in patients with metastatic melanoma [22,23,25]. An increased microbiota diversity, irrespective of species identity, was associated with improved ICI response [25,171]. Notably, baseline Faecalibacterium spp. enriched gut microbiota correlates with high peripheral blood CD4+/CD8+ effector T-cells, CD8+ TILs, and with a maintained cytokine response, and this translates into a better clinical response (PFS and OS) to ipilimumab, anti-PD-1, and ICI combination therapy in metastatic melanoma patients [23,24,25,172]. Albeit with some contrasting evidence, Bacteroides spp. seem to be associated with a reduced inflammatory response (high Treg levels, limited inflammatory cytokine concentrations) to ICIs [23,173].
Microbiota plays a role in immunomodulation through bacterial metabolites. Short-chain fatty acid (SCFA) serum levels negatively correlate with anti-CTLA-4 efficacy in metastatic melanoma patients [24]. Conversely, elevated fecal SCFAs have recently been related to better outcomes in a patient with metastatic melanoma and other solid tumors treated with anti-PD-1 therapy [26]. Notably, the composition of gut microbiota also seems to be associated with ICIs-induced colitis [23,174].

5.4. Others

As no diagnostic companions have been approved for ICI therapy in melanoma, relevant efforts have been made by the scientific and medical community to find possible biomarkers for therapy response. Among candidate biomarkers, gene-expression profiles and methylation patterns have shown possible utility in predicting therapeutic response. In pretreatment melanoma samples from patients receiving anti-PD-1 therapy, PD-L2 promoter DNA hypomethylation and high PD-L2 mRNA expression were shown to predict more prolonged progression-free survival and overall survival [175]. Gupta and colleagues reported a four-gene multiplex mRNA panel targeting PD-L1, PD-L2, CD8A, IRF1, and combined PD-L1/PD-L2 mRNA levels with promising associations with checkpoint inhibition outcomes [176]. From a recent in silico analysis of mRNA expression profiling, IRF1 and CD8A together with JAK2 and SELL have been shown to predict response to anti-PD-1 immunotherapy in melanoma patients [177]. Regarding anti-CTL4A drugs, granzyme A (GZMA) and perforin (PRF1) mRNA expression levels in pretreatment melanoma biopsies were significantly enriched in patients experiencing a clinical benefit from ipilimumab. Both transcripts have been shown to correlate with the local immune infiltrate and neoantigen load [7].
Besides TMB, mutations at specific genes seemed to predict response to ICI therapy in melanoma. Patients whose melanomas are NRAS mutated seem to respond to immunotherapy better and may have better outcomes than other genetic subtypes, suggesting that ICI therapy may be particularly effective as a treatment option for NRAS-mutant melanoma [178]. Among less frequent mutations, in two independent cohorts of patients, mutations in Serpin B3 or Serpin B4 genes have been shown to define a subgroup of patients who responded to anti-CTL4A therapy and had longer overall survival [179]. In contrast, mutations to low-density lipoprotein receptor-related protein 1B (LRP1B), as a possible surrogate biomarker for TMB, were significantly associated with a better checkpoint inhibition survival outcome [180]. Noncoding RNAs have also been investigated as possible predictive biomarkers. In a small sample cohort, higher miR-222 levels were associated with a lack of benefit to anti-CTL4 therapy [181]. In another recent study, a score of long non-coding RNAs was associated with immunotherapeutic overall survival benefit both in the IMvigor210 trial cohort (AUC, 0.79 at 12 months and 0.77 at 20 months) and in the TCGA melanoma cohort [182].

5.5. MDSCs

Myeloid-derived suppressor cells are a heterogeneous class of immature myeloid-suppressive cells [183]. Firstly, a reported strong and independently validated predictor of PFS and OS in response to anti-PD-1 was pretreatment monocytes levels rather than their gene-expression patterns or polarization (e.g., MDSCs differentiation) [57,76]. Afterward, low frequencies of MDSCs and their decline compared to baseline values were related to good prognosis and clinical benefit to ICI therapy [57,77,78,184].

5.6. ctDNA

Liquid biopsy and circulating tumor DNA (ctDNA) are increasingly integrated into clinical practice. Detectable ctDNA turned out to be associated with higher tumor burden and visceral metastases [82]. The absence of measurable ctDNA at baseline or first evaluation was an independent predictor of response and survival in PD-1 antibody-treated patients [79]. Moreover, high TMB evaluated by liquid biopsy and a >50% decrease of cell-free DNA concentration or undetectable ctDNA at three weeks after combined CTLA-4 and PD-1 antibody therapy initiation seemed to be associated with better response and OS [80].
In a recently published study exploring ctDNA evaluation, pretreatment ctDNA is a reliable and validated indicator of longer PFS for patients with metastatic melanoma in the first-line ICI treatment setting, but not in the second-line context, especially in those pretreated with BRAF/MEK inhibitors [81]. In a small cohort of metastatic melanoma patients, ctDNA assessments indicated evidence of melanoma activity that early predicted radiographic evidence of disease progression [82]. An appealing application is the longitudinal ctDNA evaluation in the challenging differentiation of pseudoprogression from real disease progression during ICI therapy. In a cohort study of 125 patients with metastatic melanoma treated with anti-PD-1 alone or in combination with ipilimumab, a reduction of at least 10-fold within the first evaluation of the number of ctDNA copies accurately identified patients with pseudoprogression. A favorable ctDNA profile, defined as undetectable baseline ctDNA that remained undetectable or detectable ctDNA at baseline that became undetectable or decreased by at least 10-fold during treatment, was significantly associated with improved OS [83]. An intriguing application is under investigation in the ongoing CAcTUs clinical trial [NCT03808441]; the longitudinal ctDNA evaluation is used to guide the switch between targeted therapy and immunotherapy in advanced cutaneous melanoma patients.
Taken together, this evidence suggests that ctDNA assessment, while not useful for “a priori” patient selection, could be an extremely promising non-invasive real-time tumor burden monitoring tool for early assessment of response to ICI therapy.

6. Conclusions

Immunotherapy has revolutionized the therapeutic landscape of melanoma. In particular, checkpoint inhibition has been shown to increase long-term outcomes, and, in some cases, it can be virtually curative. However, the absence of clinically validated predictive biomarkers is one of the major causes of the unpredictable efficacy of immunotherapy. Nevertheless, several putative predictive biomarkers have been proposed, but it seems that a single factor could not summarize the entire complexity of cancer. Indeed, none of the abovementioned biomarkers possess high sensitivity and specificity when used alone. While TMB, TILs, and PD-1 could have predictive power, it appears that a combination of multiple biomarkers could better predict the efficacy of immunotherapy. This hypothesis has culminated in the concept of a “cancer immunogram”, which accounts for several parameters that combined could estimate the efficacy of immunologic treatments [185]. In line with this concept, some interesting composite predictive tools have been developed [13,151,186,187,188]. However, in order to demonstrate an effective predictive value and a clinical utility, these composite scores should be tested prospectively. In addition, immune-related side effects might serve as an early surrogate of immunotherapy efficacy. For example, immune-induced vitiligo was shown to be linked to a favorable prognosis [189].
In perspective, liquid biopsy, ctDNA, and other circulating biomarkers need to complete development to be integrated in clinical practice with the aim to predict immunotherapy response. In addition, biomarkers might show utility also for differentiating cancer progression versus pseudoprogression.
In conclusion, it could be anticipated that in the next few years, an increasing number of useful composite predictive biomarkers tools will be available, guiding clinicians to a better stratification of patients, choice of alternative treatments to immune checkpoints, and rational de-escalation/escalation of cancer treatments. However, a deep knowledge of single biomarkers is of pivotal importance to understand why and how they should be combined.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/cancers13081819/s1, Supplementary data 1.

Author Contributions

Conceptualization, M.G.; methodology, S.B. (Serena Bonin); software, S.B. (Serena Bonin); formal analysis, S.B. (Silvia Buriolla); writing—original draft preparation, M.G., S.B. (Serena Bonin), S.B. (Silvia Buriolla), E.B.; review and editing, all the authors.; supervision, M.G. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Italian Ministry of Health (Ricerca Corrente) and by contribution “5 × 1000 per la ricerca sanitaria”.

Conflicts of Interest

M.G. reports an advisory board position for Novartis and Eli Lilly, all outside the submitted work. I.Z. reports an advisory board position for and a speaker’s fee from Sanofi Genzyme, Novartis, MSD, Sunpharma, Roche, Philogen, all outside the submitted work. F.P. reports an advisory board position for Amgen, research funding from AstraZeneca, travel grants from Celgene, research funding from an advisory board position for Eisai, an advisory board position for Eli Lilly, honoraria from Ipsen, an advisory board position for and honoraria from MSD, an advisory board position for Novartis, advisory board positions for Pierre-Fabre and Pfizer, an advisory board position for and travel grants and research funding from Roche, and honoraria from Takeda, all outside the submitted work.

Abbreviations

CIconfidence interval
ctDNAcirculating tumor DNA
ICIsimmune-checkpoint inhibitors
IHCImmunohistochemistry
LDHlactate dehydrogenase
MDSCmyeloid derived suppressor cells
MHCmajor histocompatibility complex
MMmetastatic melanoma
NGSnext-generation sequencing
NLRneutrophil to lymphocyte ratio
ORRobjective response rate
RRresponse rate
SDstandard deviation
TCRT-cell receptor
TMBtumor mutational burden

References

  1. Pollack, L.A.; Li, J.; Berkowitz, Z.; Weir, H.K.; Wu, X.-C.; Ajani, U.A.; Ekwueme, D.U.; Li, C.; Pollack, B.P. Melanoma Survival in the United States, 1992 to 2005. J. Am. Acad. Dermatol. 2011, 65, S78–S86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Ascierto, P.A.; Vecchio, M.D.; Mandalá, M.; Gogas, H.; Arance, A.M.; Dalle, S.; Cowey, C.L.; Schenker, M.; Grob, J.-J.; Chiarion-Sileni, V.; et al. Adjuvant Nivolumab versus Ipilimumab in Resected Stage IIIB–C and Stage IV Melanoma (CheckMate 238): 4-Year Results from a Multicentre, Double-Blind, Randomised, Controlled, Phase 3 Trial. Lancet Oncol. 2020, 21, 1465–1477. [Google Scholar] [CrossRef]
  3. Eggermont, A.M.M.; Blank, C.U.; Mandala, M.; Long, G.V.; Atkinson, V.; Dalle, S.; Haydon, A.; Lichinitser, M.; Khattak, A.; Carlino, M.S.; et al. Adjuvant Pembrolizumab versus Placebo in Resected Stage III Melanoma. N. Engl. J. Med. 2018, 378, 1789–1801. [Google Scholar] [CrossRef] [PubMed]
  4. Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.-J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2019, 381, 1535–1546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Couzin-Frankel, J. Cancer Immunotherapy. Science 2013, 342, 1432–1433. [Google Scholar] [CrossRef] [Green Version]
  6. Adashek, J.J.; Subbiah, I.M.; Matos, I.; Garralda, E.; Menta, A.K.; Ganeshan, D.M.; Subbiah, V. Hyperprogression and Immunotherapy: Fact, Fiction, or Alternative Fact? Trends Cancer 2020, 6, 181–191. [Google Scholar] [CrossRef] [Green Version]
  7. Allen, E.M.V.; Miao, D.; Schilling, B.; Shukla, S.A.; Blank, C.; Zimmer, L.; Sucker, A.; Hillen, U.; Foppen, M.H.G.; Goldinger, S.M.; et al. Genomic Correlates of Response to CTLA-4 Blockade in Metastatic Melanoma. Science 2015, 350, 207–211. [Google Scholar] [CrossRef] [Green Version]
  8. Snyder, A.; Makarov, V.; Merghoub, T.; Yuan, J.; Zaretsky, J.M.; Desrichard, A.; Walsh, L.A.; Postow, M.A.; Wong, P.; Ho, T.S.; et al. Genetic Basis for Clinical Response to CTLA-4 Blockade in Melanoma. N. Engl. J. Med. 2014, 371, 2189–2199. [Google Scholar] [CrossRef]
  9. Hugo, W.; Zaretsky, J.M.; Sun, L.; Song, C.; Moreno, B.H.; Hu-Lieskovan, S.; Berent-Maoz, B.; Pang, J.; Chmielowski, B.; Cherry, G.; et al. Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma. Cell 2016, 165, 35–44. [Google Scholar] [CrossRef] [Green Version]
  10. Johnson, D.B.; Frampton, G.M.; Rioth, M.J.; Yusko, E.; Xu, Y.; Guo, X.; Ennis, R.C.; Fabrizio, D.; Chalmers, Z.R.; Greenbowe, J.; et al. Targeted Next Generation Sequencing Identifies Markers of Response to PD-1 Blockade. Cancer Immunol. Res. 2016, 4, 959–967. [Google Scholar] [CrossRef] [Green Version]
  11. Weber, J.; Horak, C.; Hodi, F.S.; Chang, H.; Woods, D.; Sanders, C.; Robins, H.; Yusko, E. Baseline Tumor T Cell Receptor (TcR) Sequencing Analysis and Neo Antigen Load Is Associated with Benefit in Melanoma Patients Receiving Sequential Nivolumab and Ipilimumab. Ann. Oncol. 2016, 27, vi359. [Google Scholar] [CrossRef]
  12. Riaz, N.; Havel, J.J.; Makarov, V.; Desrichard, A.; Urba, W.J.; Sims, J.S.; Hodi, F.S.; Martín-Algarra, S.; Mandal, R.; Sharfman, W.H.; et al. Tumor and Microenvironment Evolution during Immunotherapy with Nivolumab. Cell 2017, 171, 934–949.e16. [Google Scholar] [CrossRef] [Green Version]
  13. Cristescu, R.; Mogg, R.; Ayers, M.; Albright, A.; Murphy, E.; Yearley, J.; Sher, X.; Liu, X.Q.; Lu, H.; Nebozhyn, M.; et al. Pan-Tumor Genomic Biomarkers for PD-1 Checkpoint Blockade–Based Immunotherapy. Science 2018, 362. [Google Scholar] [CrossRef] [Green Version]
  14. Roszik, J.; Haydu, L.E.; Hess, K.R.; Oba, J.; Joon, A.Y.; Siroy, A.E.; Karpinets, T.V.; Stingo, F.C.; Baladandayuthapani, V.; Tetzlaff, M.T.; et al. Novel Algorithmic Approach Predicts Tumor Mutation Load and Correlates with Immunotherapy Clinical Outcomes Using a Defined Gene Mutation Set. BMC Med. 2016, 14, 168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Roh, W.; Chen, P.-L.; Reuben, A.; Spencer, C.N.; Prieto, P.A.; Miller, J.P.; Gopalakrishnan, V.; Wang, F.; Cooper, Z.A.; Reddy, S.M.; et al. Integrated Molecular Analysis of Tumor Biopsies on Sequential CTLA-4 and PD-1 Blockade Reveals Markers of Response and Resistance. Sci. Transl. Med. 2017, 9. [Google Scholar] [CrossRef] [Green Version]
  16. Morrison, C.; Pabla, S.; Conroy, J.M.; Nesline, M.K.; Glenn, S.T.; Dressman, D.; Papanicolau-Sengos, A.; Burgher, B.; Andreas, J.; Giamo, V.; et al. Predicting Response to Checkpoint Inhibitors in Melanoma beyond PD-L1 and Mutational Burden. J. Immunother. Cancer 2018, 6, 32. [Google Scholar] [CrossRef] [PubMed]
  17. Liu, D.; Schilling, B.; Liu, D.; Sucker, A.; Livingstone, E.; Jerby-Arnon, L.; Zimmer, L.; Gutzmer, R.; Satzger, I.; Loquai, C.; et al. Integrative Molecular and Clinical Modeling of Clinical Outcomes to PD1 Blockade in Patients with Metastatic Melanoma. Nat. Med. 2019, 25, 1916–1927. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Johnson, D.B.; Estrada, M.V.; Salgado, R.; Sanchez, V.; Doxie, D.B.; Opalenik, S.R.; Vilgelm, A.E.; Feld, E.; Johnson, A.S.; Greenplate, A.R.; et al. Melanoma-Specific MHC-II Expression Represents a Tumour-Autonomous Phenotype and Predicts Response to Anti-PD-1/PD-L1 Therapy. Nat. Commun. 2016, 7, 10582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Johnson, D.B.; Bordeaux, J.; Kim, J.Y.; Vaupel, C.; Rimm, D.L.; Ho, T.H.; Joseph, R.W.; Daud, A.I.; Conry, R.M.; Gaughan, E.M.; et al. Quantitative Spatial Profiling of PD-1/PD-L1 Interaction and HLA-DR/IDO-1 Predicts Improved Outcomes of Anti-PD-1 Therapies in Metastatic Melanoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2018, 24, 5250–5260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Rodig, S.J.; Gusenleitner, D.; Jackson, D.G.; Gjini, E.; Giobbie-Hurder, A.; Jin, C.; Chang, H.; Lovitch, S.B.; Horak, C.; Weber, J.S.; et al. MHC Proteins Confer Differential Sensitivity to CTLA-4 and PD-1 Blockade in Untreated Metastatic Melanoma. Sci. Transl. Med. 2018, 10. [Google Scholar] [CrossRef] [Green Version]
  21. Sade-Feldman, M.; Jiao, Y.J.; Chen, J.H.; Rooney, M.S.; Barzily-Rokni, M.; Eliane, J.-P.; Bjorgaard, S.L.; Hammond, M.R.; Vitzthum, H.; Blackmon, S.M.; et al. Resistance to Checkpoint Blockade Therapy through Inactivation of Antigen Presentation. Nat. Commun. 2017, 8, 1136. [Google Scholar] [CrossRef]
  22. Vétizou, M.; Pitt, J.M.; Daillère, R.; Lepage, P.; Waldschmitt, N.; Flament, C.; Rusakiewicz, S.; Routy, B.; Roberti, M.P.; Duong, C.P.M.; et al. Anticancer Immunotherapy by CTLA-4 Blockade Relies on the Gut Microbiota. Science 2015, 350, 1079–1084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Chaput, N.; Lepage, P.; Coutzac, C.; Soularue, E.; Le Roux, K.; Monot, C.; Boselli, L.; Routier, E.; Cassard, L.; Collins, M.; et al. Baseline Gut Microbiota Predicts Clinical Response and Colitis in Metastatic Melanoma Patients Treated with Ipilimumab. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2017, 28, 1368–1379. [Google Scholar] [CrossRef]
  24. Coutzac, C.; Jouniaux, J.-M.; Paci, A.; Schmidt, J.; Mallardo, D.; Seck, A.; Asvatourian, V.; Cassard, L.; Saulnier, P.; Lacroix, L.; et al. Systemic Short Chain Fatty Acids Limit Antitumor Effect of CTLA-4 Blockade in Hosts with Cancer. Nat. Commun. 2020, 11, 2168. [Google Scholar] [CrossRef] [PubMed]
  25. Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut Microbiome Modulates Response to Anti-PD-1 Immunotherapy in Melanoma Patients. Science 2018, 359, 97–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Nomura, M.; Nagatomo, R.; Doi, K.; Shimizu, J.; Baba, K.; Saito, T.; Matsumoto, S.; Inoue, K.; Muto, M. Association of Short-Chain Fatty Acids in the Gut Microbiome With Clinical Response to Treatment With Nivolumab or Pembrolizumab in Patients With Solid Cancer Tumors. JAMA Netw. Open 2020, 3, e202895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Hamid, O.; Schmidt, H.; Nissan, A.; Ridolfi, L.; Aamdal, S.; Hansson, J.; Guida, M.; Hyams, D.M.; Gómez, H.; Bastholt, L.; et al. A Prospective Phase II Trial Exploring the Association between Tumor Microenvironment Biomarkers and Clinical Activity of Ipilimumab in Advanced Melanoma. J. Transl. Med. 2011, 9, 204. [Google Scholar] [CrossRef] [Green Version]
  28. Daud, A.I.; Loo, K.; Pauli, M.L.; Sanchez-Rodriguez, R.; Sandoval, P.M.; Taravati, K.; Tsai, K.; Nosrati, A.; Nardo, L.; Alvarado, M.D.; et al. Tumor Immune Profiling Predicts Response to Anti–PD-1 Therapy in Human Melanoma. J. Clin. Investig. 2016, 126, 3447–3452. [Google Scholar] [CrossRef]
  29. Tumeh, P.C.; Harview, C.L.; Yearley, J.H.; Shintaku, I.P.; Taylor, E.J.M.; Robert, L.; Chmielowski, B.; Spasic, M.; Henry, G.; Ciobanu, V.; et al. PD-1 Blockade Induces Responses by Inhibiting Adaptive Immune Resistance. Nature 2014, 515, 568–571. [Google Scholar] [CrossRef] [PubMed]
  30. Huang, A.C.; Orlowski, R.J.; Xu, X.; Mick, R.; George, S.M.; Yan, P.K.; Manne, S.; Kraya, A.A.; Wubbenhorst, B.; Dorfman, L.; et al. A Single Dose of Neoadjuvant PD-1 Blockade Predicts Clinical Outcomes in Resectable Melanoma. Nat. Med. 2019, 25, 454–461. [Google Scholar] [CrossRef]
  31. Amaria, R.N.; Reddy, S.M.; Tawbi, H.A.; Davies, M.A.; Ross, M.I.; Glitza, I.C.; Cormier, J.N.; Lewis, C.; Hwu, W.-J.; Hanna, E.; et al. Neoadjuvant Immune Checkpoint Blockade in High-Risk Resectable Melanoma. Nat. Med. 2018, 24, 1649–1654. [Google Scholar] [CrossRef]
  32. Uryvaev, A.; Passhak, M.; Hershkovits, D.; Sabo, E.; Bar-Sela, G. The Role of Tumor-Infiltrating Lymphocytes (TILs) as a Predictive Biomarker of Response to Anti-PD1 Therapy in Patients with Metastatic Non-Small Cell Lung Cancer or Metastatic Melanoma. Med. Oncol. Northwood Lond. Engl. 2018, 35, 25. [Google Scholar] [CrossRef] [PubMed]
  33. Bifulco, C.; Capone, M.; Feng, Z.; Madonna, G.; Simeone, E.; Curvietto, M.; Mozzillo, N.; Ciliberto, G.; Botti, G.; Fox, B.A.; et al. MISIPI Study: Melanoma ImmunoScore Evaluation in Patients Treated with IPIlimumab. J. Transl. Med. 2014, 12, P11. [Google Scholar] [CrossRef] [Green Version]
  34. Galon, J.; Fox, B.A.; Bifulco, C.B.; Masucci, G.; Rau, T.; Botti, G.; Marincola, F.M.; Ciliberto, G.; Pages, F.; Ascierto, P.A.; et al. Immunoscore and Immunoprofiling in Cancer: An Update from the Melanoma and Immunotherapy Bridge 2015. J. Transl. Med. 2016, 14, 273. [Google Scholar] [CrossRef] [PubMed]
  35. Karachaliou, N.; Gonzalez-Cao, M.; Crespo, G.; Drozdowskyj, A.; Aldeguer, E.; Gimenez-Capitan, A.; Teixido, C.; Molina-Vila, M.A.; Viteri, S.; De Los Llanos Gil, M.; et al. Interferon Gamma, an Important Marker of Response to Immune Checkpoint Blockade in Non-Small Cell Lung Cancer and Melanoma Patients. Ther. Adv. Med. Oncol. 2018, 10, 1758834017749748. [Google Scholar] [CrossRef] [PubMed]
  36. Ayers, M.; Lunceford, J.; Nebozhyn, M.; Murphy, E.; Loboda, A.; Kaufman, D.R.; Albright, A.; Cheng, J.D.; Kang, S.P.; Shankaran, V.; et al. IFN-γ-Related MRNA Profile Predicts Clinical Response to PD-1 Blockade. J. Clin. Investig. 2017, 127, 2930–2940. [Google Scholar] [CrossRef] [PubMed]
  37. Chen, P.-L.; Roh, W.; Reuben, A.; Cooper, Z.A.; Spencer, C.N.; Prieto, P.A.; Miller, J.P.; Bassett, R.L.; Gopalakrishnan, V.; Wani, K.; et al. Analysis of Immune Signatures in Longitudinal Tumor Samples Yields Insight into Biomarkers of Response and Mechanisms of Resistance to Immune Checkpoint Blockade. Cancer Discov. 2016, 6, 827–837. [Google Scholar] [CrossRef] [Green Version]
  38. Gide, T.N.; Quek, C.; Menzies, A.M.; Tasker, A.T.; Shang, P.; Holst, J.; Madore, J.; Lim, S.Y.; Velickovic, R.; Wongchenko, M.; et al. Distinct Immune Cell Populations Define Response to Anti-PD-1 Monotherapy and Anti-PD-1/Anti-CTLA-4 Combined Therapy. Cancer Cell 2019, 35, 238–255.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Diem, S.; Kasenda, B.; Spain, L.; Martin-Liberal, J.; Marconcini, R.; Gore, M.; Larkin, J. Serum Lactate Dehydrogenase as an Early Marker for Outcome in Patients Treated with Anti-PD-1 Therapy in Metastatic Melanoma. Br. J. Cancer 2016, 114, 256–261. [Google Scholar] [CrossRef] [PubMed]
  40. Wagner, N.B.; Forschner, A.; Leiter, U.; Garbe, C.; Eigentler, T.K. S100B and LDH as Early Prognostic Markers for Response and Overall Survival in Melanoma Patients Treated with Anti-PD-1 or Combined Anti-PD-1 plus Anti-CTLA-4 Antibodies. Br. J. Cancer 2018, 119, 339–346. [Google Scholar] [CrossRef] [PubMed]
  41. Simeone, E.; Gentilcore, G.; Giannarelli, D.; Grimaldi, A.M.; Caracò, C.; Curvietto, M.; Esposito, A.; Paone, M.; Palla, M.; Cavalcanti, E.; et al. Immunological and Biological Changes during Ipilimumab Treatment and Their Potential Correlation with Clinical Response and Survival in Patients with Advanced Melanoma. Cancer Immunol. Immunother. 2014, 63, 675–683. [Google Scholar] [CrossRef] [PubMed]
  42. Laino, A.S.; Woods, D.; Vassallo, M.; Qian, X.; Tang, H.; Wind-Rotolo, M.; Weber, J. Serum Interleukin-6 and C-Reactive Protein Are Associated with Survival in Melanoma Patients Receiving Immune Checkpoint Inhibition. J. Immunother. Cancer 2020, 8. [Google Scholar] [CrossRef]
  43. Wolchok, J.D.; Chiarion-Sileni, V.; Gonzalez, R.; Rutkowski, P.; Grob, J.-J.; Cowey, C.L.; Lao, C.D.; Wagstaff, J.; Schadendorf, D.; Ferrucci, P.F.; et al. Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2017, 377, 1345–1356. [Google Scholar] [CrossRef] [PubMed]
  44. Dupuis, F.; Lamant, L.; Gerard, E.; Torossian, N.; Chaltiel, L.; Filleron, T.; Beylot-Barry, M.; Dutriaux, C.; Prey, S.; Gros, A.; et al. Clinical, Histological and Molecular Predictors of Metastatic Melanoma Responses to Anti-PD-1 Immunotherapy. Br. J. Cancer 2018, 119, 193–199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Long, G.V.; Atkinson, V.; Cebon, J.S.; Jameson, M.B.; Fitzharris, B.M.; McNeil, C.M.; Hill, A.G.; Ribas, A.; Atkins, M.B.; Thompson, J.A.; et al. Standard-Dose Pembrolizumab in Combination with Reduced-Dose Ipilimumab for Patients with Advanced Melanoma (KEYNOTE-029): An Open-Label, Phase 1b Trial. Lancet Oncol. 2017, 18, 1202–1210. [Google Scholar] [CrossRef] [Green Version]
  46. Pérottet, J.; Le Goff, E.; Legoupil, D.; Quéré, G.; Schick, U.; Marcorelles, P.; Uguen, A. PD-L1 Copy Number Variation Does Not Correlate With PD-L1 Expression or Response to Anti-PD-1 Immunotherapy In Patients With Advanced Melanomas. Appl. Immunohistochem. Mol. Morphol. AIMM 2020, 28, 161–165. [Google Scholar] [CrossRef]
  47. Puzanov, I.; Dummer, R.; Schachter, J.; Pavlick, A.C.; Gonzalez, R.; Ascierto, P.A.; Margolin, K.A.; Hamid, O.; Agarwala, S.S.; Carlino, M.S.; et al. Efficacy Based on Tumor PD-L1 Expression in KEYNOTE-002, a Randomized Comparison of Pembrolizumab (Pembro; MK-3475) versus Chemotherapy in Patients (Pts) with Ipilimumab-Refractory (IPI-R) Advanced Melanoma (MEL). J. Clin. Oncol. 2015, 33, 3012. [Google Scholar] [CrossRef]
  48. Valpione, S.; Pasquali, S.; Campana, L.G.; Piccin, L.; Mocellin, S.; Pigozzo, J.; Chiarion-Sileni, V. Sex and Interleukin-6 Are Prognostic Factors for Autoimmune Toxicity Following Treatment with Anti-CTLA4 Blockade. J. Transl. Med. 2018, 16, 94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Sanmamed, M.F.; Perez-Gracia, J.L.; Schalper, K.A.; Fusco, J.P.; Gonzalez, A.; Rodriguez-Ruiz, M.E.; Oñate, C.; Perez, G.; Alfaro, C.; Martín-Algarra, S.; et al. Changes in Serum Interleukin-8 (IL-8) Levels Reflect and Predict Response to Anti-PD-1 Treatment in Melanoma and Non-Small-Cell Lung Cancer Patients. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2017, 28, 1988–1995. [Google Scholar] [CrossRef] [PubMed]
  50. Fujimura, T.; Sato, Y.; Tanita, K.; Kambayashi, Y.; Otsuka, A.; Fujisawa, Y.; Yoshino, K.; Matsushita, S.; Funakoshi, T.; Hata, H.; et al. Serum Levels of Soluble CD163 and CXCL5 May Be Predictive Markers for Immune-Related Adverse Events in Patients with Advanced Melanoma Treated with Nivolumab: A Pilot Study. Oncotarget 2018, 9, 15542–15551. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Fujimura, T.; Sato, Y.; Tanita, K.; Lyu, C.; Kambayashi, Y.; Amagai, R.; Otsuka, A.; Fujisawa, Y.; Yoshino, K.; Matsushita, S.; et al. Association of Baseline Serum Levels of CXCL5 With the Efficacy of Nivolumab in Advanced Melanoma. Front. Med. 2019, 6, 86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Dronca, R.S.; Mansfield, A.S.; Liu, X.; Harrington, S.; Enninga, E.A.; Kottschade, L.A.; Koo, C.W.; McWilliams, R.R.; Block, M.S.; Nevala, W.K.; et al. Bim and Soluble PD-L1 (SPD-L1) as Predictive Biomarkers of Response to Anti-PD-1 Therapy in Patients with Melanoma and Lung Carcinoma. J. Clin. Oncol. 2017, 35, 11534. [Google Scholar] [CrossRef]
  53. Zhou, J.; Mahoney, K.M.; Giobbie-Hurder, A.; Zhao, F.; Lee, S.; Liao, X.; Rodig, S.; Li, J.; Wu, X.; Butterfield, L.H.; et al. Soluble PD-L1 as a Biomarker in Malignant Melanoma Treated with Checkpoint Blockade. Cancer Immunol. Res. 2017, 5, 480–492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Tucci, M.; Passarelli, A.; Mannavola, F.; Stucci, L.S.; Ascierto, P.A.; Capone, M.; Madonna, G.; Lopalco, P.; Silvestris, F. Serum Exosomes as Predictors of Clinical Response to Ipilimumab in Metastatic Melanoma. Oncoimmunology 2018, 7, e1387706. [Google Scholar] [CrossRef]
  55. Cordonnier, M.; Nardin, C.; Chanteloup, G.; Derangere, V.; Algros, M.-P.; Arnould, L.; Garrido, C.; Aubin, F.; Gobbo, J. Tracking the Evolution of Circulating Exosomal-PD-L1 to Monitor Melanoma Patients. J. Extracell. Vesicles 2020, 9, 1710899. [Google Scholar] [CrossRef] [Green Version]
  56. Pistillo, M.P.; Fontana, V.; Morabito, A.; Dozin, B.; Laurent, S.; Carosio, R.; Banelli, B.; Ferrero, F.; Spano, L.; Tanda, E.; et al. Soluble CTLA-4 as a Favorable Predictive Biomarker in Metastatic Melanoma Patients Treated with Ipilimumab: An Italian Melanoma Intergroup Study. Cancer Immunol. Immunother. 2019, 68, 97–107. [Google Scholar] [CrossRef]
  57. Weide, B.; Martens, A.; Hassel, J.C.; Berking, C.; Postow, M.A.; Bisschop, K.; Simeone, E.; Mangana, J.; Schilling, B.; Di Giacomo, A.M.; et al. Baseline Biomarkers for Outcome of Melanoma Patients Treated with Pembrolizumab. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2016, 22, 5487–5496. [Google Scholar] [CrossRef] [Green Version]
  58. Delyon, J.; Mateus, C.; Lefeuvre, D.; Lanoy, E.; Zitvogel, L.; Chaput, N.; Roy, S.; Eggermont, A.M.M.; Routier, E.; Robert, C. Experience in Daily Practice with Ipilimumab for the Treatment of Patients with Metastatic Melanoma: An Early Increase in Lymphocyte and Eosinophil Counts Is Associated with Improved Survival. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2013, 24, 1697–1703. [Google Scholar] [CrossRef]
  59. Martens, A.; Wistuba-Hamprecht, K.; Geukes Foppen, M.; Yuan, J.; Postow, M.A.; Wong, P.; Romano, E.; Khammari, A.; Dreno, B.; Capone, M.; et al. Baseline Peripheral Blood Biomarkers Associated with Clinical Outcome of Advanced Melanoma Patients Treated with Ipilimumab. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2016, 22, 2908–2918. [Google Scholar] [CrossRef] [Green Version]
  60. Nakamura, Y.; Kitano, S.; Takahashi, A.; Tsutsumida, A.; Namikawa, K.; Tanese, K.; Abe, T.; Funakoshi, T.; Yamamoto, N.; Amagai, M.; et al. Nivolumab for Advanced Melanoma: Pretreatment Prognostic Factors and Early Outcome Markers during Therapy. Oncotarget 2016, 7, 77404–77415. [Google Scholar] [CrossRef]
  61. Ferrucci, P.F.; Gandini, S.; Battaglia, A.; Alfieri, S.; Di Giacomo, A.M.; Giannarelli, D.; Cappellini, G.C.A.; De Galitiis, F.; Marchetti, P.; Amato, G.; et al. Baseline Neutrophil-to-Lymphocyte Ratio Is Associated with Outcome of Ipilimumab-Treated Metastatic Melanoma Patients. Br. J. Cancer 2015, 112, 1904–1910. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Ferrucci, P.F.; Ascierto, P.A.; Pigozzo, J.; Del Vecchio, M.; Maio, M.; Antonini Cappellini, G.C.; Guidoboni, M.; Queirolo, P.; Savoia, P.; Mandalà, M.; et al. Baseline Neutrophils and Derived Neutrophil-to-Lymphocyte Ratio: Prognostic Relevance in Metastatic Melanoma Patients Receiving Ipilimumab. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2016, 27, 732–738. [Google Scholar] [CrossRef]
  63. Capone, M.; Giannarelli, D.; Mallardo, D.; Madonna, G.; Festino, L.; Grimaldi, A.M.; Vanella, V.; Simeone, E.; Paone, M.; Palmieri, G.; et al. Baseline Neutrophil-to-Lymphocyte Ratio (NLR) and Derived NLR Could Predict Overall Survival in Patients with Advanced Melanoma Treated with Nivolumab. J. Immunother. Cancer 2018, 6, 74. [Google Scholar] [CrossRef] [Green Version]
  64. Bartlett, E.K.; Flynn, J.R.; Panageas, K.S.; Ferraro, R.A.; Sta Cruz, J.M.; Postow, M.A.; Coit, D.G.; Ariyan, C.E. High Neutrophil-to-Lymphocyte Ratio (NLR) Is Associated with Treatment Failure and Death in Patients Who Have Melanoma Treated with PD-1 Inhibitor Monotherapy. Cancer 2020, 126, 76–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Zaragoza, J.; Caille, A.; Beneton, N.; Bens, G.; Christiann, F.; Maillard, H.; Machet, L. High Neutrophil to Lymphocyte Ratio Measured before Starting Ipilimumab Treatment Is Associated with Reduced Overall Survival in Patients with Melanoma. Br. J. Dermatol. 2016, 174, 146–151. [Google Scholar] [CrossRef] [PubMed]
  66. Cassidy, M.R.; Wolchok, R.E.; Zheng, J.; Panageas, K.S.; Wolchok, J.D.; Coit, D.; Postow, M.A.; Ariyan, C. Neutrophil to Lymphocyte Ratio Is Associated With Outcome During Ipilimumab Treatment. EBioMedicine 2017, 18, 56–61. [Google Scholar] [CrossRef] [Green Version]
  67. Fujisawa, Y.; Yoshino, K.; Otsuka, A.; Funakoshi, T.; Fujimura, T.; Yamamoto, Y.; Hata, H.; Tanaka, R.; Yamaguchi, K.; Nonomura, Y.; et al. Baseline Neutrophil to Lymphocyte Ratio Combined with Serum Lactate Dehydrogenase Level Associated with Outcome of Nivolumab Immunotherapy in a Japanese Advanced Melanoma Population. Br. J. Dermatol. 2018, 179, 213–215. [Google Scholar] [CrossRef] [PubMed]
  68. Wistuba-Hamprecht, K.; Martens, A.; Heubach, F.; Romano, E.; Geukes Foppen, M.; Yuan, J.; Postow, M.; Wong, P.; Mallardo, D.; Schilling, B.; et al. Peripheral CD8 Effector-Memory Type 1 T-Cells Correlate with Outcome in Ipilimumab-Treated Stage IV Melanoma Patients. Eur. J. Cancer Oxf. Engl. 1990 2017, 73, 61–70. [Google Scholar] [CrossRef] [Green Version]
  69. Tietze, J.K.; Angelova, D.; Heppt, M.V.; Ruzicka, T.; Berking, C. Low Baseline Levels of NK Cells May Predict a Positive Response to Ipilimumab in Melanoma Therapy. Exp. Dermatol. 2017, 26, 622–629. [Google Scholar] [CrossRef] [PubMed]
  70. Weber, J.S.; Kudchadkar, R.R.; Yu, B.; Gallenstein, D.; Horak, C.E.; Inzunza, H.D.; Zhao, X.; Martinez, A.J.; Wang, W.; Gibney, G.; et al. Safety, Efficacy, and Biomarkers of Nivolumab with Vaccine in Ipilimumab-Refractory or -Naive Melanoma. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2013, 31, 4311–4318. [Google Scholar] [CrossRef] [Green Version]
  71. Bochem, J.; Zelba, H.; Amaral, T.; Spreuer, J.; Soffel, D.; Eigentler, T.; Wagner, N.B.; Uslu, U.; Terheyden, P.; Meier, F.; et al. Peripheral PD-1+CD56+ T-Cell Frequencies Correlate with Outcome in Stage IV Melanoma under PD-1 Blockade. PLoS ONE 2019, 14, e0221301. [Google Scholar] [CrossRef] [PubMed]
  72. Subrahmanyam, P.B.; Dong, Z.; Gusenleitner, D.; Giobbie-Hurder, A.; Severgnini, M.; Zhou, J.; Manos, M.; Eastman, L.M.; Maecker, H.T.; Hodi, F.S. Distinct Predictive Biomarker Candidates for Response to Anti-CTLA-4 and Anti-PD-1 Immunotherapy in Melanoma Patients. J. Immunother. Cancer 2018, 6, 18. [Google Scholar] [CrossRef] [PubMed]
  73. Pirozyan, M.R.; McGuire, H.M.; Emran, A.A.; Tseng, H.-Y.; Tiffen, J.C.; Lee, J.H.; Carlino, M.S.; Menzies, A.M.; Long, G.V.; Scolyer, R.A.; et al. Pretreatment Innate Cell Populations and CD4 T Cells in Blood Are Associated With Response to Immune Checkpoint Blockade in Melanoma Patients. Front. Immunol. 2020, 11, 372. [Google Scholar] [CrossRef] [PubMed]
  74. Cha, E.; Klinger, M.; Hou, Y.; Cummings, C.; Ribas, A.; Faham, M.; Fong, L. Improved Survival with T Cell Clonotype Stability after Anti-CTLA-4 Treatment in Cancer Patients. Sci. Transl. Med. 2014, 6, 238ra70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Postow, M.A.; Chesney, J.; Pavlick, A.C.; Robert, C.; Grossmann, K.; McDermott, D.; Linette, G.P.; Meyer, N.; Giguere, J.K.; Agarwala, S.S.; et al. Nivolumab and Ipilimumab versus Ipilimumab in Untreated Melanoma. N. Engl. J. Med. 2015, 372, 2006–2017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Krieg, C.; Nowicka, M.; Guglietta, S.; Schindler, S.; Hartmann, F.J.; Weber, L.M.; Dummer, R.; Robinson, M.D.; Levesque, M.P.; Becher, B. High-Dimensional Single-Cell Analysis Predicts Response to Anti-PD-1 Immunotherapy. Nat. Med. 2018, 24, 144–153. [Google Scholar] [CrossRef]
  77. de Coaña, Y.P.; Wolodarski, M.; Poschke, I.; Yoshimoto, Y.; Yang, Y.; Nyström, M.; Edbäck, U.; Brage, S.E.; Lundqvist, A.; Masucci, G.V.; et al. Ipilimumab Treatment Decreases Monocytic MDSCs and Increases CD8 Effector Memory T Cells in Long-Term Survivors with Advanced Melanoma. Oncotarget 2017, 8, 21539–21553. [Google Scholar] [CrossRef] [Green Version]
  78. Meyer, C.; Cagnon, L.; Costa-Nunes, C.M.; Baumgaertner, P.; Montandon, N.; Leyvraz, L.; Michielin, O.; Romano, E.; Speiser, D.E. Frequencies of Circulating MDSC Correlate with Clinical Outcome of Melanoma Patients Treated with Ipilimumab. Cancer Immunol. Immunother. 2014, 63, 247–257. [Google Scholar] [CrossRef] [Green Version]
  79. Lee, J.H.; Long, G.V.; Boyd, S.; Lo, S.; Menzies, A.M.; Tembe, V.; Guminski, A.; Jakrot, V.; Scolyer, R.A.; Mann, G.J.; et al. Circulating Tumour DNA Predicts Response to Anti-PD1 Antibodies in Metastatic Melanoma. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2017, 28, 1130–1136. [Google Scholar] [CrossRef]
  80. Forschner, A.; Battke, F.; Hadaschik, D.; Schulze, M.; Weißgraeber, S.; Han, C.-T.; Kopp, M.; Frick, M.; Klumpp, B.; Tietze, N.; et al. Tumor Mutation Burden and Circulating Tumor DNA in Combined CTLA-4 and PD-1 Antibody Therapy in Metastatic Melanoma - Results of a Prospective Biomarker Study. J. Immunother. Cancer 2019, 7, 180. [Google Scholar] [CrossRef]
  81. Marsavela, G.; Lee, J.; Calapre, L.; Wong, S.Q.; Pereira, M.R.; McEvoy, A.C.; Reid, A.L.; Robinson, C.; Warburton, L.; Abed, A.; et al. Circulating Tumor DNA Predicts Outcome from First-, but Not Second-Line Treatment and Identifies Melanoma Patients Who May Benefit from Combination Immunotherapy. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2020, 26, 5926–5933. [Google Scholar] [CrossRef] [PubMed]
  82. Rowe, S.P.; Luber, B.; Makell, M.; Brothers, P.; Santmyer, J.; Schollenberger, M.D.; Quinn, H.; Edelstein, D.L.; Jones, F.S.; Bleich, K.B.; et al. From Validity to Clinical Utility: The Influence of Circulating Tumor DNA on Melanoma Patient Management in a Real-World Setting. Mol. Oncol. 2018, 12, 1661–1672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Lee, J.H.; Long, G.V.; Menzies, A.M.; Lo, S.; Guminski, A.; Whitbourne, K.; Peranec, M.; Scolyer, R.; Kefford, R.F.; Rizos, H.; et al. Association Between Circulating Tumor DNA and Pseudoprogression in Patients With Metastatic Melanoma Treated With Anti-Programmed Cell Death 1 Antibodies. JAMA Oncol. 2018, 4, 717–721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Galuppini, F.; Dal Pozzo, C.A.; Deckert, J.; Loupakis, F.; Fassan, M.; Baffa, R. Tumor Mutation Burden: From Comprehensive Mutational Screening to the Clinic. Cancer Cell Int. 2019, 19, 209. [Google Scholar] [CrossRef] [PubMed]
  85. Meléndez, B.; Van Campenhout, C.; Rorive, S.; Remmelink, M.; Salmon, I.; D’Haene, N. Methods of Measurement for Tumor Mutational Burden in Tumor Tissue. Transl. Lung Cancer Res. 2018, 7, 661–667. [Google Scholar] [CrossRef]
  86. Vilimas, T. Measuring Tumor Mutational Burden Using Whole-Exome Sequencing. In Biomarkers for Immunotherapy of Cancer; Humana: New York, NY, USA, 2020; Volume 2055, pp. 63–91. [Google Scholar] [CrossRef]
  87. Campesato, L.F.; Barroso-Sousa, R.; Jimenez, L.; Correa, B.R.; Sabbaga, J.; Hoff, P.M.; Reis, L.F.L.; Galante, P.A.F.; Camargo, A.A. Comprehensive Cancer-Gene Panels Can Be Used to Estimate Mutational Load and Predict Clinical Benefit to PD-1 Blockade in Clinical Practice. Oncotarget 2015, 6, 34221–34227. [Google Scholar] [CrossRef] [Green Version]
  88. Chalmers, Z.R.; Connelly, C.F.; Fabrizio, D.; Gay, L.; Ali, S.M.; Ennis, R.; Schrock, A.; Campbell, B.; Shlien, A.; Chmielecki, J.; et al. Analysis of 100,000 Human Cancer Genomes Reveals the Landscape of Tumor Mutational Burden. Genome Med. 2017, 9, 34. [Google Scholar] [CrossRef]
  89. Samstein, R.M.; Lee, C.-H.; Shoushtari, A.N.; Hellmann, M.D.; Shen, R.; Janjigian, Y.Y.; Barron, D.A.; Zehir, A.; Jordan, E.J.; Omuro, A.; et al. Tumor Mutational Load Predicts Survival after Immunotherapy across Multiple Cancer Types. Nat. Genet. 2019, 51, 202–206. [Google Scholar] [CrossRef]
  90. Davis, A.A.; Chae, Y.K.; Agte, S.; Pan, A.; Simon, N.I.; Taxter, T.J.; Behdad, A.; Carneiro, B.A.; Cristofanilli, M.; Giles, F.J. Comparison of Tumor Mutational Burden (TMB) across Tumor Tissue and Circulating Tumor DNA (CtDNA). J. Clin. Oncol. 2017, 35, e23028. [Google Scholar] [CrossRef]
  91. Peters, S.; Cho, B.C.; Reinmuth, N.; Lee, K.H.; Luft, A.; Ahn, M.-J.; Baas, P.; Dols, M.C.; Smolin, A.; Vicente, D.; et al. Abstract CT074: Tumor Mutational Burden (TMB) as a Biomarker of Survival in Metastatic Non-Small Cell Lung Cancer (MNSCLC): Blood and Tissue TMB Analysis from MYSTIC, a Phase III Study of First-Line Durvalumab ± Tremelimumab vs Chemotherapy. Cancer Res. 2019, 79, CT074. [Google Scholar] [CrossRef]
  92. Wang, Z.; Duan, J.; Cai, S.; Han, M.; Dong, H.; Zhao, J.; Zhu, B.; Wang, S.; Zhuo, M.; Sun, J.; et al. Assessment of Blood Tumor Mutational Burden as a Potential Biomarker for Immunotherapy in Patients With Non-Small Cell Lung Cancer With Use of a Next-Generation Sequencing Cancer Gene Panel. JAMA Oncol. 2019, 5, 696–702. [Google Scholar] [CrossRef] [PubMed]
  93. Gandara, D.R.; Paul, S.M.; Kowanetz, M.; Schleifman, E.; Zou, W.; Li, Y.; Rittmeyer, A.; Fehrenbacher, L.; Otto, G.; Malboeuf, C.; et al. Blood-Based Tumor Mutational Burden as a Predictor of Clinical Benefit in Non-Small-Cell Lung Cancer Patients Treated with Atezolizumab. Nat. Med. 2018, 24, 1441–1448. [Google Scholar] [CrossRef] [PubMed]
  94. Vokes, N.I.; Liu, D.; Ricciuti, B.; Jimenez-Aguilar, E.; Rizvi, H.; Dietlein, F.; He, M.X.; Margolis, C.A.; Elmarakeby, H.A.; Girshman, J.; et al. Harmonization of Tumor Mutational Burden Quantification and Association With Response to Immune Checkpoint Blockade in Non–Small-Cell Lung Cancer. JCO Precis. Oncol. 2019, 1–12. [Google Scholar] [CrossRef] [PubMed]
  95. Yarchoan, M.; Hopkins, A.; Jaffee, E.M. Tumor Mutational Burden and Response Rate to PD-1 Inhibition. N. Engl. J. Med. 2017, 377, 2500–2501. [Google Scholar] [CrossRef] [PubMed]
  96. TMB Harmonization Working Group Meeting | Friends of Cancer Research. Available online: https://friendsofcancerresearch.org/events/tmb-harmonization-working-group-meeting (accessed on 4 November 2020).
  97. Goodman, A.M.; Kato, S.; Bazhenova, L.; Patel, S.P.; Frampton, G.M.; Miller, V.; Stephens, P.J.; Daniels, G.A.; Kurzrock, R. Tumor Mutational Burden as an Independent Predictor of Response to Immunotherapy in Diverse Cancers. Mol. Cancer Ther. 2017, 16, 2598–2608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Garrido, F.; Ruiz-Cabello, F.; Aptsiauri, N. Rejection versus Escape: The Tumor MHC Dilemma. Cancer Immunol. Immunother. 2017, 66, 259–271. [Google Scholar] [CrossRef]
  99. Axelrod, M.L.; Cook, R.S.; Johnson, D.B.; Balko, J.M. Biological Consequences of MHC-II Expression by Tumor Cells in Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019, 25, 2392–2402. [Google Scholar] [CrossRef]
  100. Farhood, B.; Najafi, M.; Mortezaee, K. CD8+ Cytotoxic T Lymphocytes in Cancer Immunotherapy: A Review. J. Cell. Physiol. 2019, 234, 8509–8521. [Google Scholar] [CrossRef]
  101. Tay, R.E.; Richardson, E.K.; Toh, H.C. Revisiting the Role of CD4 + T Cells in Cancer Immunotherapy—New Insights into Old Paradigms. Cancer Gene Ther. 2020, 1–13. [Google Scholar] [CrossRef]
  102. Olbryt, M.; Rajczykowski, M.; Widłak, W. Biological Factors behind Melanoma Response to Immune Checkpoint Inhibitors. Int. J. Mol. Sci. 2020, 21, 4071. [Google Scholar] [CrossRef]
  103. Merelli, B.; Massi, D.; Cattaneo, L.; Mandalà, M. Targeting the PD1/PD-L1 Axis in Melanoma: Biological Rationale, Clinical Challenges and Opportunities. Crit. Rev. Oncol. Hematol. 2014, 89, 140–165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Yarchoan, M.; Albacker, L.A.; Hopkins, A.C.; Montesion, M.; Murugesan, K.; Vithayathil, T.T.; Zaidi, N.; Azad, N.S.; Laheru, D.A.; Frampton, G.M.; et al. PD-L1 Expression and Tumor Mutational Burden Are Independent Biomarkers in Most Cancers. JCI Insight 2019, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Emancipator, K. Keytruda and PD-L1: A Real-World Example of Co-Development of a Drug with a Predictive Biomarker. AAPS J. 2020, 23, 5. [Google Scholar] [CrossRef] [PubMed]
  106. FDA; U.F.; D.A. Companion Diagnostics. Available online: https://www.fda.gov/medical-devices/vitro-diagnostics/companion-diagnostics (accessed on 29 October 2020).
  107. Krigsfeld, G.S.; Prince, E.A.; Pratt, J.; Chizhevsky, V.; William Ragheb, J.; Novotny, J.; Huron, D. Analysis of Real-World PD-L1 IHC 28-8 and 22C3 PharmDx Assay Utilisation, Turnaround Times and Analytical Concordance across Multiple Tumour Types. J. Clin. Pathol. 2020, 73, 656–664. [Google Scholar] [CrossRef] [PubMed]
  108. O’Malley, D.P.; Yang, Y.; Boisot, S.; Sudarsanam, S.; Wang, J.-F.; Chizhevsky, V.; Zhao, G.; Arain, S.; Weiss, L.M. Immunohistochemical Detection of PD-L1 among Diverse Human Neoplasms in a Reference Laboratory: Observations Based upon 62,896 Cases. Mod. Pathol. 2019, 32, 929–942. [Google Scholar] [CrossRef] [PubMed]
  109. Daud, A.I.; Wolchok, J.D.; Robert, C.; Hwu, W.-J.; Weber, J.S.; Ribas, A.; Hodi, F.S.; Joshua, A.M.; Kefford, R.; Hersey, P.; et al. Programmed Death-Ligand 1 Expression and Response to the Anti-Programmed Death 1 Antibody Pembrolizumab in Melanoma. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2016, 34, 4102–4109. [Google Scholar] [CrossRef] [PubMed]
  110. Carbognin, L.; Pilotto, S.; Milella, M.; Vaccaro, V.; Brunelli, M.; Caliò, A.; Cuppone, F.; Sperduti, I.; Giannarelli, D.; Chilosi, M.; et al. Differential Activity of Nivolumab, Pembrolizumab and MPDL3280A According to the Tumor Expression of Programmed Death-Ligand-1 (PD-L1): Sensitivity Analysis of Trials in Melanoma, Lung and Genitourinary Cancers. PLoS ONE 2015, 10, e0130142. [Google Scholar] [CrossRef] [Green Version]
  111. Yamazaki, N.; Takenouchi, T.; Fujimoto, M.; Ihn, H.; Uchi, H.; Inozume, T.; Kiyohara, Y.; Uhara, H.; Nakagawa, K.; Furukawa, H.; et al. Phase 1b Study of Pembrolizumab (MK-3475; Anti-PD-1 Monoclonal Antibody) in Japanese Patients with Advanced Melanoma (KEYNOTE-041). Cancer Chemother. Pharmacol. 2017, 79, 651–660. [Google Scholar] [CrossRef] [Green Version]
  112. Madonna, G.; Ballesteros-Merino, C.; Feng, Z.; Bifulco, C.; Capone, M.; Giannarelli, D.; Mallardo, D.; Simeone, E.; Grimaldi, A.M.; Caracò, C.; et al. PD-L1 Expression with Immune-Infiltrate Evaluation and Outcome Prediction in Melanoma Patients Treated with Ipilimumab. Oncoimmunology 2018, 7, e1405206. [Google Scholar] [CrossRef] [Green Version]
  113. Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B.; et al. Safety, Activity, and Immune Correlates of Anti-PD-1 Antibody in Cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef]
  114. Robert, C.; Long, G.V.; Brady, B.; Dutriaux, C.; Maio, M.; Mortier, L.; Hassel, J.C.; Rutkowski, P.; McNeil, C.; Kalinka-Warzocha, E.; et al. Nivolumab in Previously Untreated Melanoma without BRAF Mutation. N. Engl. J. Med. 2015, 372, 320–330. [Google Scholar] [CrossRef] [Green Version]
  115. Cho, J.; Ahn, S.; Yoo, K.H.; Kim, J.H.; Choi, S.-H.; Jang, K.-T.; Lee, J. Treatment Outcome of PD-1 Immune Checkpoint Inhibitor in Asian Metastatic Melanoma Patients: Correlative Analysis with PD-L1 Immunohistochemistry. Investig. New Drugs 2016, 34, 677–684. [Google Scholar] [CrossRef]
  116. Weber, J.S.; D’Angelo, S.P.; Minor, D.; Hodi, F.S.; Gutzmer, R.; Neyns, B.; Hoeller, C.; Khushalani, N.I.; Miller, W.H.; Lao, C.D.; et al. Nivolumab versus Chemotherapy in Patients with Advanced Melanoma Who Progressed after Anti-CTLA-4 Treatment (CheckMate 037): A Randomised, Controlled, Open-Label, Phase 3 Trial. Lancet Oncol. 2015, 16, 375–384. [Google Scholar] [CrossRef]
  117. Larkin, J.; Lao, C.D.; Urba, W.J.; McDermott, D.F.; Horak, C.; Jiang, J.; Wolchok, J.D. Efficacy and Safety of Nivolumab in Patients With BRAF V600 Mutant and BRAF Wild-Type Advanced Melanoma: A Pooled Analysis of 4 Clinical Trials. JAMA Oncol. 2015, 1, 433–440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Daud, A.; Hamid, O.; Robert, C.; Hodi, F.S.; Wolchok, J.D.; Hwu, W.J.; Weber, J.S.; Kefford, R.; Hersey, P.; Joshua, A.M.; et al. Relationship between Programmed Death Ligand 1 (PD-L1) Expression and Clinical Outcome in Patients (Pts) with Melanoma (MEL) Treated with Pembrolizumab (Pembro; MK-3475). Eur. J. Cancer 2014, 50, 48–49. [Google Scholar] [CrossRef]
  119. Long, G.V.; Dummer, R.; Hamid, O.; Gajewski, T.F.; Caglevic, C.; Dalle, S.; Arance, A.; Carlino, M.S.; Grob, J.-J.; Kim, T.M.; et al. Epacadostat plus Pembrolizumab versus Placebo plus Pembrolizumab in Patients with Unresectable or Metastatic Melanoma (ECHO-301/KEYNOTE-252): A Phase 3, Randomised, Double-Blind Study. Lancet Oncol. 2019, 20, 1083–1097. [Google Scholar] [CrossRef]
  120. Robert, C.; Ribas, A.; Schachter, J.; Arance, A.; Grob, J.-J.; Mortier, L.; Daud, A.; Carlino, M.S.; McNeil, C.M.; Lotem, M.; et al. Pembrolizumab versus Ipilimumab in Advanced Melanoma (KEYNOTE-006): Post-Hoc 5-Year Results from an Open-Label, Multicentre, Randomised, Controlled, Phase 3 Study. Lancet Oncol. 2019, 20, 1239–1251. [Google Scholar] [CrossRef]
  121. Robert, C.; Ribas, A.; Hamid, O.; Daud, A.; Wolchok, J.D.; Joshua, A.M.; Hwu, W.-J.; Weber, J.S.; Gangadhar, T.C.; Joseph, R.W.; et al. Durable Complete Response After Discontinuation of Pembrolizumab in Patients With Metastatic Melanoma. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2018, 36, 1668–1674. [Google Scholar] [CrossRef]
  122. Diggs, L.P.; Hsueh, E.C. Utility of PD-L1 Immunohistochemistry Assays for Predicting PD-1/PD-L1 Inhibitor Response. Biomark. Res. 2017, 5, 12. [Google Scholar] [CrossRef] [Green Version]
  123. Gibney, G.T.; Weiner, L.M.; Atkins, M.B. Predictive Biomarkers for Checkpoint Inhibitor-Based Immunotherapy. Lancet Oncol. 2016, 17, e542–e551. [Google Scholar] [CrossRef] [Green Version]
  124. Mantovani, A.; Allavena, P.; Sica, A.; Balkwill, F. Cancer-Related Inflammation. Nature 2008, 454, 436–444. [Google Scholar] [CrossRef]
  125. Carlino, M.S.; Long, G.V.; Schadendorf, D.; Robert, C.; Ribas, A.; Richtig, E.; Nyakas, M.; Caglevic, C.; Tarhini, A.; Blank, C.; et al. Outcomes by Line of Therapy and Programmed Death Ligand 1 Expression in Patients with Advanced Melanoma Treated with Pembrolizumab or Ipilimumab in KEYNOTE-006: A Randomised Clinical Trial. Eur. J. Cancer Oxf. Engl. 1990 2018, 101, 236–243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Clark, W.H.; From, L.; Bernardino, E.A.; Mihm, M.C. The Histogenesis and Biologic Behavior of Primary Human Malignant Melanomas of the Skin. Cancer Res. 1969, 29, 705–727. [Google Scholar] [PubMed]
  127. Clemente, C.G.; Mihm, M.C.; Bufalino, R.; Zurrida, S.; Collini, P.; Cascinelli, N. Prognostic Value of Tumor Infiltrating Lymphocytes in the Vertical Growth Phase of Primary Cutaneous Melanoma. Cancer 1996, 77, 1303–1310. [Google Scholar] [CrossRef]
  128. Taylor, R.C.; Patel, A.; Panageas, K.S.; Busam, K.J.; Brady, M.S. Tumor-Infiltrating Lymphocytes Predict Sentinel Lymph Node Positivity in Patients with Cutaneous Melanoma. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2007, 25, 869–875. [Google Scholar] [CrossRef]
  129. Tuthill, R.J.; Unger, J.M.; Liu, P.Y.; Flaherty, L.E.; Sondak, V.K. Southwest Oncology Group Risk Assessment in Localized Primary Cutaneous Melanoma: A Southwest Oncology Group Study Evaluating Nine Factors and a Test of the Clark Logistic Regression Prediction Model. Am. J. Clin. Pathol. 2002, 118, 504–511. [Google Scholar] [CrossRef] [PubMed]
  130. Mlecnik, B.; Bindea, G.; Pagès, F.; Galon, J. Tumor Immunosurveillance in Human Cancers. Cancer Metastasis Rev. 2011, 30, 5–12. [Google Scholar] [CrossRef] [Green Version]
  131. Weiss, S.A.; Han, S.W.; Lui, K.; Tchack, J.; Shapiro, R.; Berman, R.; Zhong, J.; Krogsgaard, M.; Osman, I.; Darvishian, F. Immunologic Heterogeneity of Tumor-Infiltrating Lymphocyte Composition in Primary Melanoma. Hum. Pathol. 2016, 57, 116–125. [Google Scholar] [CrossRef] [Green Version]
  132. Antohe, M.; Nedelcu, R.I.; Nichita, L.; Popp, C.G.; Cioplea, M.; Brinzea, A.; Hodorogea, A.; Calinescu, A.; Balaban, M.; Ion, D.A.; et al. Tumor Infiltrating Lymphocytes: The Regulator of Melanoma Evolution. Oncol. Lett. 2019, 17, 4155–4161. [Google Scholar] [CrossRef]
  133. Ascierto, P.A.; Capone, M.; Urba, W.J.; Bifulco, C.B.; Botti, G.; Lugli, A.; Marincola, F.M.; Ciliberto, G.; Galon, J.; Fox, B.A. The Additional Facet of Immunoscore: Immunoprofiling as a Possible Predictive Tool for Cancer Treatment. J. Transl. Med. 2013, 11, 54. [Google Scholar] [CrossRef]
  134. Nanda, V.G.Y.; Peng, W.; Hwu, P.; Davies, M.A.; Ciliberto, G.; Fattore, L.; Malpicci, D.; Aurisicchio, L.; Ascierto, P.A.; Croce, C.M.; et al. Melanoma and Immunotherapy Bridge 2015. J. Transl. Med. 2016, 14, 65. [Google Scholar] [CrossRef] [Green Version]
  135. Spranger, S.; Spaapen, R.M.; Zha, Y.; Williams, J.; Meng, Y.; Ha, T.T.; Gajewski, T.F. Up-Regulation of PD-L1, IDO, and T(Regs) in the Melanoma Tumor Microenvironment Is Driven by CD8(+) T Cells. Sci. Transl. Med. 2013, 5, 200ra116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Meireson, A.; Devos, M.; Brochez, L. IDO Expression in Cancer: Different Compartment, Different Functionality? Front. Immunol. 2020, 11, 531491. [Google Scholar] [CrossRef] [PubMed]
  137. Radvanyi, L.G. Tumor-Infiltrating Lymphocyte Therapy: Addressing Prevailing Questions. Cancer J. Sudbury Mass 2015, 21, 450–464. [Google Scholar] [CrossRef] [PubMed]
  138. Mastracci, L.; Fontana, V.; Queirolo, P.; Carosio, R.; Grillo, F.; Morabito, A.; Banelli, B.; Tanda, E.; Boutros, A.; Dozin, B.; et al. Response to Ipilimumab Therapy in Metastatic Melanoma Patients: Potential Relevance of CTLA-4+ Tumor Infiltrating Lymphocytes and Their in Situ Localization. Cancer Immunol. Immunother. 2020, 69, 653–662. [Google Scholar] [CrossRef] [PubMed]
  139. Oble, D.A.; Loewe, R.; Yu, P.; Mihm, M.C. Focus on TILs: Prognostic significance of tumor infiltrating lymphocytes in human melanoma. Cancer Immun. 2009, 9, 3. [Google Scholar]
  140. Parigi, S.M.; Czarnewski, P.; Das, S.; Steeg, C.; Brockmann, L.; Fernandez-Gaitero, S.; Yman, V.; Forkel, M.; Höög, C.; Mjösberg, J.; et al. Flt3 Ligand Expands Bona Fide Innate Lymphoid Cell Precursors in Vivo. Sci. Rep. 2018, 8, 154. [Google Scholar] [CrossRef]
  141. Barry, K.C.; Hsu, J.; Broz, M.L.; Cueto, F.J.; Binnewies, M.; Combes, A.J.; Nelson, A.E.; Loo, K.; Kumar, R.; Rosenblum, M.D.; et al. A Natural Killer-Dendritic Cell Axis Defines Checkpoint Therapy-Responsive Tumor Microenvironments. Nat. Med. 2018, 24, 1178–1191. [Google Scholar] [CrossRef]
  142. de Andrade, L.F.; Lu, Y.; Luoma, A.; Ito, Y.; Pan, D.; Pyrdol, J.W.; Yoon, C.H.; Yuan, G.-C.; Wucherpfennig, K.W. Discovery of Specialized NK Cell Populations Infiltrating Human Melanoma Metastases. JCI Insight 2019, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Cassetta, L.; Kitamura, T. Targeting Tumor-Associated Macrophages as a Potential Strategy to Enhance the Response to Immune Checkpoint Inhibitors. Front. Cell Dev. Biol. 2018, 6, 38. [Google Scholar] [CrossRef]
  144. Cassetta, L.; Kitamura, T. Macrophage Targeting: Opening New Possibilities for Cancer Immunotherapy. Immunology 2018, 155, 285–293. [Google Scholar] [CrossRef] [PubMed]
  145. Kim, J.M.; Chen, D.S. Immune Escape to PD-L1/PD-1 Blockade: Seven Steps to Success (or Failure). Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2016, 27, 1492–1504. [Google Scholar] [CrossRef] [PubMed]
  146. Galon, J.; Angell, H.K.; Bedognetti, D.; Marincola, F.M. The Continuum of Cancer Immunosurveillance: Prognostic, Predictive, and Mechanistic Signatures. Immunity 2013, 39, 11–26. [Google Scholar] [CrossRef] [Green Version]
  147. Sobhani, N.; Corona, S.P.; Roviello, G.; Bagby, S.; D’Angelo, A.; Iezzi, G.; Generali, D. Immune-Gene Signature: A New Tool for Patient Selection for Checkpoint Inhibitors? Future Oncol. Lond. Engl. 2020, 16, 1327–1330. [Google Scholar] [CrossRef]
  148. Rozeman, E.A.; Menzies, A.M.; van Akkooi, A.C.J.; Adhikari, C.; Bierman, C.; van de Wiel, B.A.; Scolyer, R.A.; Krijgsman, O.; Sikorska, K.; Eriksson, H.; et al. Identification of the Optimal Combination Dosing Schedule of Neoadjuvant Ipilimumab plus Nivolumab in Macroscopic Stage III Melanoma (OpACIN-Neo): A Multicentre, Phase 2, Randomised, Controlled Trial. Lancet Oncol. 2019, 20, 948–960. [Google Scholar] [CrossRef]
  149. Blank, C.U.; Rozeman, E.A.; Fanchi, L.F.; Sikorska, K.; van de Wiel, B.; Kvistborg, P.; Krijgsman, O.; van den Braber, M.; Philips, D.; Broeks, A.; et al. Neoadjuvant versus Adjuvant Ipilimumab plus Nivolumab in Macroscopic Stage III Melanoma. Nat. Med. 2018, 24, 1655–1661. [Google Scholar] [CrossRef] [PubMed]
  150. Ji, R.-R.; Chasalow, S.D.; Wang, L.; Hamid, O.; Schmidt, H.; Cogswell, J.; Alaparthy, S.; Berman, D.; Jure-Kunkel, M.; Siemers, N.O.; et al. An Immune-Active Tumor Microenvironment Favors Clinical Response to Ipilimumab. Cancer Immunol. Immunother. 2012, 61, 1019–1031. [Google Scholar] [CrossRef]
  151. Jiang, P.; Gu, S.; Pan, D.; Fu, J.; Sahu, A.; Hu, X.; Li, Z.; Traugh, N.; Bu, X.; Li, B.; et al. Signatures of T Cell Dysfunction and Exclusion Predict Cancer Immunotherapy Response. Nat. Med. 2018, 24, 1550–1558. [Google Scholar] [CrossRef] [PubMed]
  152. Petrelli, F.; Ardito, R.; Merelli, B.; Lonati, V.; Cabiddu, M.; Seghezzi, S.; Barni, S.; Ghidini, A. Prognostic and Predictive Role of Elevated Lactate Dehydrogenase in Patients with Melanoma Treated with Immunotherapy and BRAF Inhibitors: A Systematic Review and Meta-Analysis. Melanoma Res. 2019, 29, 1–12. [Google Scholar] [CrossRef]
  153. Balch, C.M.; Gershenwald, J.E.; Soong, S.-J.; Thompson, J.F.; Atkins, M.B.; Byrd, D.R.; Buzaid, A.C.; Cochran, A.J.; Coit, D.G.; Ding, S.; et al. Final Version of 2009 AJCC Melanoma Staging and Classification. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2009, 27, 6199–6206. [Google Scholar] [CrossRef] [Green Version]
  154. Gershenwald, J.E.; Scolyer, R.A.; Hess, K.R.; Sondak, V.K.; Long, G.V.; Ross, M.I.; Lazar, A.J.; Faries, M.B.; Kirkwood, J.M.; McArthur, G.A.; et al. Melanoma Staging: Evidence-Based Changes in the American Joint Committee on Cancer Eighth Edition Cancer Staging Manual. CA Cancer J. Clin. 2017, 67, 472–492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Van Wilpe, S.; Koornstra, R.; Den Brok, M.; De Groot, J.W.; Blank, C.; De Vries, J.; Gerritsen, W.; Mehra, N. Lactate Dehydrogenase: A Marker of Diminished Antitumor Immunity. Oncoimmunology 2020, 9, 1731942. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Singer, K.; Gottfried, E.; Kreutz, M.; Mackensen, A. Suppression of T-Cell Responses by Tumor Metabolites. Cancer Immunol. Immunother. 2011, 60, 425–431. [Google Scholar] [CrossRef] [PubMed]
  157. Hauschild, A.; Engel, G.; Brenner, W.; Gläser, R.; Mönig, H.; Henze, E.; Christophers, E. S100B Protein Detection in Serum Is a Significant Prognostic Factor in Metastatic Melanoma. Oncology 1999, 56, 338–344. [Google Scholar] [CrossRef] [PubMed]
  158. Harpio, R.; Einarsson, R. S100 Proteins as Cancer Biomarkers with Focus on S100B in Malignant Melanoma. Clin. Biochem. 2004, 37, 512–518. [Google Scholar] [CrossRef]
  159. Fang, S.; Wang, Y.; Sui, D.; Liu, H.; Ross, M.I.; Gershenwald, J.E.; Cormier, J.N.; Royal, R.E.; Lucci, A.; Schacherer, C.W.; et al. C-Reactive Protein as a Marker of Melanoma Progression. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2015, 33, 1389–1396. [Google Scholar] [CrossRef] [PubMed]
  160. Nyakas, M.; Aamdal, E.; Jacobsen, K.D.; Guren, T.K.; Aamdal, S.; Hagene, K.T.; Brunsvig, P.; Yndestad, A.; Halvorsen, B.; Tasken, K.A.; et al. Prognostic Biomarkers for Immunotherapy with Ipilimumab in Metastatic Melanoma. Clin. Exp. Immunol. 2019, 197, 74–82. [Google Scholar] [CrossRef] [Green Version]
  161. Weber, J.S.; Tang, H.; Hippeli, L.; Qian, M.; Wind-Rotolo, M.; Larkin, J.M.G.; Wolchok, J.D.; Sznol, M.; Robert, C.; Woods, D.M.; et al. Serum IL-6 and CRP as Prognostic Factors in Melanoma Patients Receiving Single Agent and Combination Checkpoint Inhibition. J. Clin. Oncol. 2019, 37, 100. [Google Scholar] [CrossRef]
  162. Kubo, Y.; Fukushima, S.; Inamori, Y.; Tsuruta, M.; Egashira, S.; Yamada-Kanazawa, S.; Nakahara, S.; Tokuzumi, A.; Miyashita, A.; Aoi, J.; et al. Serum Concentrations of HGF Are Correlated with Response to Anti-PD-1 Antibody Therapy in Patients with Metastatic Melanoma. J. Dermatol. Sci. 2019, 93, 33–40. [Google Scholar] [CrossRef] [Green Version]
  163. Tobin, R.P.; Jordan, K.R.; Kapoor, P.; Spongberg, E.; Davis, D.; Vorwald, V.M.; Couts, K.L.; Gao, D.; Smith, D.E.; Borgers, J.S.W.; et al. IL-6 and IL-8 Are Linked With Myeloid-Derived Suppressor Cell Accumulation and Correlate With Poor Clinical Outcomes in Melanoma Patients. Front. Oncol. 2019, 9, 1223. [Google Scholar] [CrossRef] [Green Version]
  164. Chen, S.-H.; Gong, X.; Zhang, Y.; Van Horn, R.D.; Yin, T.; Huber, L.; Burke, T.F.; Manro, J.; Iversen, P.W.; Wu, W.; et al. RAF Inhibitor LY3009120 Sensitizes RAS or BRAF Mutant Cancer to CDK4/6 Inhibition by Abemaciclib via Superior Inhibition of Phospho-RB and Suppression of Cyclin D1. Oncogene 2018, 37, 821–832. [Google Scholar] [CrossRef] [PubMed]
  165. Martens, A.; Wistuba-Hamprecht, K.; Yuan, J.; Postow, M.A.; Wong, P.; Capone, M.; Madonna, G.; Khammari, A.; Schilling, B.; Sucker, A.; et al. Increases in Absolute Lymphocytes and Circulating CD4+ and CD8+ T Cells Are Associated with Positive Clinical Outcome of Melanoma Patients Treated with Ipilimumab. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2016, 22, 4848–4858. [Google Scholar] [CrossRef] [Green Version]
  166. Capone, M.; Fratangelo, F.; Giannarelli, D.; Sorrentino, C.; Turiello, R.; Zanotta, S.; Galati, D.; Madonna, G.; Tuffanelli, M.; Scarpato, L.; et al. Frequency of Circulating CD8+CD73+T Cells Is Associated with Survival in Nivolumab-Treated Melanoma Patients. J. Transl. Med. 2020, 18, 121. [Google Scholar] [CrossRef] [Green Version]
  167. Sen, D.R.; Kaminski, J.; Barnitz, R.A.; Kurachi, M.; Gerdemann, U.; Yates, K.B.; Tsao, H.-W.; Godec, J.; LaFleur, M.W.; Brown, F.D.; et al. The Epigenetic Landscape of T Cell Exhaustion. Science 2016, 354, 1165–1169. [Google Scholar] [CrossRef] [Green Version]
  168. Fuertes Marraco, S.A.; Neubert, N.J.; Verdeil, G.; Speiser, D.E. Inhibitory Receptors Beyond T Cell Exhaustion. Front. Immunol. 2015, 6, 310. [Google Scholar] [CrossRef]
  169. Postow, M.A.; Manuel, M.; Wong, P.; Yuan, J.; Dong, Z.; Liu, C.; Perez, S.; Tanneau, I.; Noel, M.; Courtier, A.; et al. Peripheral T Cell Receptor Diversity Is Associated with Clinical Outcomes Following Ipilimumab Treatment in Metastatic Melanoma. J. Immunother. Cancer 2015, 3, 23. [Google Scholar] [CrossRef] [Green Version]
  170. Thursby, E.; Juge, N. Introduction to the Human Gut Microbiota. Biochem. J. 2017, 474, 1823–1836. [Google Scholar] [CrossRef]
  171. Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C.P.M.; Alou, M.T.; Daillère, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P.; et al. Gut Microbiome Influences Efficacy of PD-1-Based Immunotherapy against Epithelial Tumors. Science 2018, 359, 91–97. [Google Scholar] [CrossRef] [Green Version]
  172. Frankel, A.E.; Coughlin, L.A.; Kim, J.; Froehlich, T.W.; Xie, Y.; Frenkel, E.P.; Koh, A.Y. Metagenomic Shotgun Sequencing and Unbiased Metabolomic Profiling Identify Specific Human Gut Microbiota and Metabolites Associated with Immune Checkpoint Therapy Efficacy in Melanoma Patients. Neoplasia 2017, 19, 848–855. [Google Scholar] [CrossRef]
  173. Peters, B.A.; Wilson, M.; Moran, U.; Pavlick, A.; Izsak, A.; Wechter, T.; Weber, J.S.; Osman, I.; Ahn, J. Relating the Gut Metagenome and Metatranscriptome to Immunotherapy Responses in Melanoma Patients. Genome Med. 2019, 11, 61. [Google Scholar] [CrossRef] [Green Version]
  174. Dubin, K.; Callahan, M.K.; Ren, B.; Khanin, R.; Viale, A.; Ling, L.; No, D.; Gobourne, A.; Littmann, E.; Huttenhower, C.; et al. Intestinal Microbiome Analyses Identify Melanoma Patients at Risk for Checkpoint-Blockade-Induced Colitis. Nat. Commun. 2016, 7, 10391. [Google Scholar] [CrossRef] [Green Version]
  175. Hoffmann, F.; Zarbl, R.; Niebel, D.; Sirokay, J.; Fröhlich, A.; Posch, C.; Holderried, T.A.W.; Brossart, P.; Saavedra, G.; Kuster, P.; et al. Prognostic and Predictive Value of PD-L2 DNA Methylation and MRNA Expression in Melanoma. Clin. Epigenetics 2020, 12, 94. [Google Scholar] [CrossRef]
  176. Gupta, S.; McCann, L.; Chan, Y.G.Y.; Lai, E.W.; Wei, W.; Wong, P.F.; Smithy, J.W.; Weidler, J.; Rhees, B.; Bates, M.; et al. Closed System RT-QPCR as a Potential Companion Diagnostic Test for Immunotherapy Outcome in Metastatic Melanoma. J. Immunother. Cancer 2019, 7, 254. [Google Scholar] [CrossRef]
  177. Wang, X.; Chai, Z.; Li, Y.; Long, F.; Hao, Y.; Pan, G.; Liu, M.; Li, B. Identification of Potential Biomarkers for Anti-PD-1 Therapy in Melanoma by Weighted Correlation Network Analysis. Genes 2020, 11, 435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Johnson, D.B.; Lovly, C.M.; Flavin, M.; Panageas, K.S.; Ayers, G.D.; Zhao, Z.; Iams, W.T.; Colgan, M.; DeNoble, S.; Terry, C.R.; et al. Impact of NRAS Mutations for Patients with Advanced Melanoma Treated with Immune Therapies. Cancer Immunol. Res. 2015, 3, 288–295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Riaz, N.; Havel, J.J.; Kendall, S.M.; Makarov, V.; Walsh, L.A.; Desrichard, A.; Weinhold, N.; Chan, T.A. Recurrent SERPINB3 and SERPINB4 Mutations in Patients Who Respond to Anti-CTLA4 Immunotherapy. Nat. Genet. 2016, 48, 1327–1329. [Google Scholar] [CrossRef]
  180. Chen, H.; Chong, W.; Wu, Q.; Yao, Y.; Mao, M.; Wang, X. Association of LRP1B Mutation With Tumor Mutation Burden and Outcomes in Melanoma and Non-Small Cell Lung Cancer Patients Treated With Immune Check-Point Blockades. Front. Immunol. 2019, 10, 1113. [Google Scholar] [CrossRef] [Green Version]
  181. Galore-Haskel, G.; Nemlich, Y.; Greenberg, E.; Ashkenazi, S.; Hakim, M.; Itzhaki, O.; Shoshani, N.; Shapira-Fromer, R.; Ben-Ami, E.; Ofek, E.; et al. A Novel Immune Resistance Mechanism of Melanoma Cells Controlled by the ADAR1 Enzyme. Oncotarget 2015, 6, 28999–29015. [Google Scholar] [CrossRef] [Green Version]
  182. Yu, Y.; Zhang, W.; Li, A.; Chen, Y.; Ou, Q.; He, Z.; Zhang, Y.; Liu, R.; Yao, H.; Song, E. Association of Long Noncoding RNA Biomarkers With Clinical Immune Subtype and Prediction of Immunotherapy Response in Patients With Cancer. JAMA Netw. Open 2020, 3, e202149. [Google Scholar] [CrossRef] [Green Version]
  183. Bronte, V.; Brandau, S.; Chen, S.-H.; Colombo, M.P.; Frey, A.B.; Greten, T.F.; Mandruzzato, S.; Murray, P.J.; Ochoa, A.; Ostrand-Rosenberg, S.; et al. Recommendations for Myeloid-Derived Suppressor Cell Nomenclature and Characterization Standards. Nat. Commun. 2016, 7, 12150. [Google Scholar] [CrossRef] [Green Version]
  184. Weber, J.; Gibney, G.; Kudchadkar, R.; Yu, B.; Cheng, P.; Martinez, A.J.; Kroeger, J.; Richards, A.; McCormick, L.; Moberg, V.; et al. Phase I/II Study of Metastatic Melanoma Patients Treated with Nivolumab Who Had Progressed after Ipilimumab. Cancer Immunol. Res. 2016, 4, 345–353. [Google Scholar] [CrossRef] [Green Version]
  185. Blank, C.U.; Haanen, J.B.; Ribas, A.; Schumacher, T.N. CANCER IMMUNOLOGY. The “Cancer Immunogram.”. Science 2016, 352, 658–660. [Google Scholar] [CrossRef] [PubMed]
  186. Anagnostou, V.; Niknafs, N.; Marrone, K.; Bruhm, D.C.; White, J.R.; Naidoo, J.; Hummelink, K.; Monkhorst, K.; Lalezari, F.; Lanis, M.; et al. Multimodal Genomic Features Predict Outcome of Immune Checkpoint Blockade in Non-Small-Cell Lung Cancer. Nat. Cancer 2020, 1, 99–111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Nabet, B.Y.; Esfahani, M.S.; Moding, E.J.; Hamilton, E.G.; Chabon, J.J.; Rizvi, H.; Steen, C.B.; Chaudhuri, A.A.; Liu, C.L.; Hui, A.B.; et al. Noninvasive Early Identification of Therapeutic Benefit from Immune Checkpoint Inhibition. Cell 2020, 183, 363–376.e13. [Google Scholar] [CrossRef]
  188. Auslander, N.; Zhang, G.; Lee, J.S.; Frederick, D.T.; Miao, B.; Moll, T.; Tian, T.; Wei, Z.; Madan, S.; Sullivan, R.J.; et al. Robust Prediction of Response to Immune Checkpoint Blockade Therapy in Metastatic Melanoma. Nat. Med. 2018, 24, 1545–1549. [Google Scholar] [CrossRef] [PubMed]
  189. Teulings, H.-E.; Limpens, J.; Jansen, S.N.; Zwinderman, A.H.; Reitsma, J.B.; Spuls, P.I.; Luiten, R.M. Vitiligo-Like Depigmentation in Patients With Stage III-IV Melanoma Receiving Immunotherapy and Its Association With Survival: A Systematic Review and Meta-Analysis. J. Clin. Oncol. 2015, 33, 773–781. [Google Scholar] [CrossRef]
Figure 1. Forest plots of odds ratio (in log scale) of response to ICI (checkpoint inhibitor) in metastatic or non-resectable melanoma patients according to positivity to PD-L1 by immunohistochemistry. Meta-analysis was sub-grouped according to ICI treatment. The size of squares mirrors the weight of the study in the meta-analysis.
Figure 1. Forest plots of odds ratio (in log scale) of response to ICI (checkpoint inhibitor) in metastatic or non-resectable melanoma patients according to positivity to PD-L1 by immunohistochemistry. Meta-analysis was sub-grouped according to ICI treatment. The size of squares mirrors the weight of the study in the meta-analysis.
Cancers 13 01819 g001
Table 1. List of predictive biomarkers for immunotherapy in melanoma.
Table 1. List of predictive biomarkers for immunotherapy in melanoma.
BiomarkerStudy Details & ReferencePatientsClinical Utility
TMB
(WES)
Ipilimumab
Van Allen et al. [7]
110CB *
TMB
(WES)
Ipilimumab or tremelimumab
Snyder et al. [8]
64OS
TMB
(WES)
Nivolumab or pembrolizumab
Hugo et al. [9]
38OS
TMB
(NGS)
Nivolumab or pembrolizumab or atezolizumab
Johnson et al. [10]
65RR, PFS, OS
TMB
(WES)
Nivolumab and ipilimumab
Weber et al. [11]
94RR for the sequence:
Nivo → Ipi
TMB
(WES)
Nivolumab
Riaz et al. [12]
68OS
TMB
(WES)
Pembrolizumab
Cristescu et al. [13]
89RR
TMB
(NGS)
Ipilimumab or adoptive T-cell therapy
Rosizik et al. [14]
76PFS, OS
TMB
(WES)
Anti-PD-1 or anti-CTLA-4
Roh et al. [15]
21RR (numerical)
TMB
(NGS)
Anti-PD-1 and/or anti-CTLA-4
Morrison et al. [16]
160RR
MHC-IINivolumab or pembrolizumab
Liu et al. [17]
32RR
(in previously exposed to anti-CTLA-4)
MHC-IIAnti-PD-1
Johnson et al. [18]
30RR, PFS, OS
MHC-IINivolumab or pembrolizumab
Johnson et al. [19]
166RR, PFS
MHC-I
MHC-II
Ipilimumab and/or nivolumab
Rodig et al. [20]
181OS
(MHC-I for ipilimumab;
MHC-II for nivolumab)
B2MAnti-CTLA-4 or anti-PD-1
Sade-Feldman et al. [21]
143RR, OS
Gut MicrobiotaAnti-CTLA-4
Vetizou et al. [22]
25Antitumor response
Gut MicrobiotaAnti-CTLA-4
Chaput et al. [23]
26PFS, OS
Gut MicrobiotaAnti-CTLA-4
Coutzac et al. [24]
85PFS, OS
Gut microbiotaanti-PD-1
Gopalakrishnan et al. [25]
112ORR, PFS
Gut microbiota Anti-PD-1
Nomura et al. [26]
52ORR, PFS
TILsAnti-CTLA-4
Hamid et al. [27]
82RR
TILsAnti-PD-1
Daud et al. [28]
20RR, PFS
TILsAnti-PD-1
Tumeh et al. [29]
46RR
TILsAnti-PD-1
Huang et al. [30]
27PCR, DFS
TILsAnti-PD-1 or Anti-PD-1 + anti-CTLA-4
Amaria et al. [31]
23ORR
TILsAnti-PD-1
Uryvaev et al. [32]
30RR, PFS
ImmunoscoreAnti-CTLA-4
Bifulco et al., Galon et al. [33,34]
190No relationship with RR
IFN signatureAnti-PD-1
Karachaliou et al. [35]
21DFS, PFS, OS
IFN signatureAnti-PD-1
Ayers et al. [36]
19RR, PFS
Expanded immune signatureAnti-PD-1
Ayers et al. [36]
62RR, PFS
Immune signatureAnti-PD-1
Chen et al. [37]
46RR
Immune signatureAnti-PD-1
Huang et al. [30]
27RFS
Immune signatureAnti-PD-1
Gide et al. [38]
158 (samples)PFS
LDH Anti-PD-1
Diem et al. [39]
66OS, ORR
LDHAnti PD-1 or anti-CTLA-4
Wagner et al. [40]
238OS
LDHAnti-CTLA-4
Simeone et al. [41]
95OS, RR
S100BAnti PD-1 or anti-CTLA-4
Wagner et al. [40]
238OS
PCRAnti-PD-1 end/or anti-CTLA-4
Laino et al. [42]
Wagner et al. [40]
1503OS, RR
PCRAnti-CTLA-4
Simeone et al. [43]
95OS, RR
PCRAnti-CTLA-4
Nyakas et al. [44]
69OS
HGFAnti-PD-1
Kubo et al. [45]
29OS, PFS
IL-6Anti-PD-1 end/or anti-CTLA-4
Laino et al. [46]
Wagner et al. [47]
1503OS, RR
IL-6Anti-CTLA-4
Valpione et al. [48]
140OS
IL-8Anti-PD-1 ± anti-CTL-4
Sanmamed et al. [49]
29 + 34 validation cohort NSCLC + melanoma pts)OS, RR
CXCL-5Anti-PD-1
Fujimura et al. [50,51]
46ORR
sPDL-1Anti-PD-1
Dronca et al. [52]
60ORR
sPDL-1Anti-CTLA-4 or anti-PDL-1
Zhou et al. [53]
90ORR
PD-L1 and CD28 exosomal expressionAnti-CTLA-4
Tucci et al. [54]
59CR, PFS, OS
Exosomal PD-L1 Anti-PDL-1 and/or anti-CTLA-4
Cordonnier et al. [55]
100RR, PFS, OS
CTLA-4Anti-CTLA-4
Pistillo et al. [56]
113ORR
Absolute lymphocyte countAnti-CTLA-4
Simeone et al. [41]
95OS, RR
Relative eosinophil and lymphocyte count Anti-PDL-1
Weide et al. [57]
616OS
Eosinophil and lymphocyte count Anti-CTLA-4
Delyon et al. [58]
73OS
Absolute lymphocyte countAnti-CTLA-4
Martens et al. [59]
82OS
Absolute monocyte/eosinophil and relative lymphocyte countsAnti-CTLA-4
Martens et al. [59]
209OS, RR
Absolute lymphocyte and neutrophil countAnti-PDL-1
Nakamura et al. [60]
98OS
NLRAnti-PD-1 end/or anti-CTLA-4
Laino et al. [42]
1503OS, RR
NLRAnti-CTLA-4
Ferrucci et al. [61]
69PFS, OS
NLRAnti-CTLA-4
Ferrucci et al. [62]
720PFS, OS
NLRAnti-PD-1
Capone et al. [63]
97PFS, OS
NLRAnti-PD-1
Bartlett et al. [64]
224TTF, OS
NLRAnti-CTLA-4
Zaragoza et al. [65]
58OS
NLRAnti-CTLA-4
Cassidy et al. [66]
197RR, PFS, OS
NLRAnti-PD-1
Fujisawa et al. [67]
90ORR
CD8 effector-memory type 1 T-cellsAnti-CTLA-4
Wistuba-Hamprecht et al. [68]
137OS, RR
CD45RO+CD8+ T-cells Anti-CTLA-4 or anti-PD-1
Tietze et al. [69]
30OS
FoxP3-TregsAnti-CTLA-4
Simeone et al. [41]
95OS, RR
FoxP3-TregsAnti-CTLA-4
Martens et al. [59]
209OS, RR
FoxP3-TregsNivolumab with vaccine
Weber et al. [70]
90ORR
CD37Anti-PD-1
Capone et al. [63]
100ORR, OS
PD-1+CD56+ T-cellsAnti-PD-1
Bochem et al. [71]
75PFS, OS, RR
CD4 and CD8 memory T- cellsAnti-CTLA-4
Martens et al. [59]
82OS
NK T-cellsAnti-PD-1
Subrahmanyam et al. [72]
67ORR
T-cell exhaustion markersAnti-PD-1
Pirozyan et al. [73]
42No differences in RR
TCR repertoireNivolumab with vaccine
Weber et al. [70]
90ORR
TCR repertoireAnti-CTLA-4
Cha et al. [74]
21 melanoma ptsORR
TCR repertoireAnti-CTLA-4
Postow et al. [75]
12CB *
Monocyte frequencyAnti-PD-1
Weide et al. [57]
616OS
Monocyte frequencyAnti-PD-1
Krieg et al. [76]
60 samplesPFS, OS
MDSCSsAnti-PD-1
Weide et al. [57]
616OS
MDSCSsAnti-PD-1
De Coana et al. [77]
43ORR, OS
MDSCSsAnti-CTLA-4
Meyer et al. [78]
49
(+15 controls)
ORR
MDSCSsAnti-CTLA-4
Weber et al. [11]
92ORR, PFS, OS
ctDNAAnti-PD-1 (± anti-CTLA-4)
Lee et al. [79]
86RR, PFS, OS
TMB, ctDNA, cell-free DNAAnti-PD-1 or anti-CTLA-4
Forschner et al. [80]
35ORR, OS
ctDNAAnti-PD-1
Marsavela et al. [81]
125 patients discovery cohort + 128 validation samplesPFS, OS
ctDNAAnti-PD-L1 and/or anti-CTLA-4 or BRAF/MEK inhibitors
Rowe et al. [82]
127ctDNA mutant fraction and sum of tumor diameters relationship
ctDNAAnti-PD-1 (± anti-CTLA-4)
Lee et al. [83]
125ORR, OS
CB: clinical benefit; ctDNA: circulating tumor DNA; Ipi: ipilimumab; MDSCSs: myeloid derived suppressor cells; NGS: next-generation sequencing; Nivo: nivolumab; NLR: neutrophil to lymphocyte ratio; OS: overall survival; TMB: tumor mutational burden. * defined as evidence of tumor burden reduction or prolonged stable disease lasting at least 9 months.
Table 2. ORR in PD-L1-positive and -negative strafied by treatment type.
Table 2. ORR in PD-L1-positive and -negative strafied by treatment type.
ReferencesTreatmentMean ORR in PD-L1-Positive (SD, 95% CI)Mean ORR in PD-L1-Negative (SD, 95% CI)p * Value
[43,46,47,110,111,118,119,121,125]Pembrolizumab41.3
(23.2, 24.8–57.9)
23.0
(17.7, 10.4–35.7)
0.009
[75,112,118,125]Ipilimumab22.6
(11.5, 4.4–40.9)
10.0
(4.3, 3.2–16.8)
0.1
[43,46,70,110,113,116,117,120]Nivolumab47.5
(9.6, 40.1–54.9)
24.0
(19.5, 8.9–39.0)
0.01
[43,118]Nivolumab + ipilimumab65.2
(9.7, -22.0–152.4)
55.5
(0.3, 53.3–57.8)
0.4
§ [16,19,44,45,110,114,115]Combined therapy and atezolizumab (1 study)46.2
(19.8, 29.6–62.7)
27.7
(18.6, 12.1–43.2)
0.0008
Overall results based on a total of 5069 patients44.1
(19.0, 37.3–50.9)
24.5
(18.5, 18.0–31.0)
<0.0001
§ Treatments include combinations of nivolumab and pembrolizumab (2 studies); ipilimumab and pembrolizumab (1 study); treatment with nivolumab or ipilimumab or their combination (1 study); nivolumab or pembrolizumab (2 studies); atezolizumab (1 study), nivolumab and other treatments (1 study). * t-test for paired data. ORR: objective response rate; SD: standard deviation; CI: confidence interval.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Garutti, M.; Bonin, S.; Buriolla, S.; Bertoli, E.; Pizzichetta, M.A.; Zalaudek, I.; Puglisi, F. Find the Flame: Predictive Biomarkers for Immunotherapy in Melanoma. Cancers 2021, 13, 1819. https://doi.org/10.3390/cancers13081819

AMA Style

Garutti M, Bonin S, Buriolla S, Bertoli E, Pizzichetta MA, Zalaudek I, Puglisi F. Find the Flame: Predictive Biomarkers for Immunotherapy in Melanoma. Cancers. 2021; 13(8):1819. https://doi.org/10.3390/cancers13081819

Chicago/Turabian Style

Garutti, Mattia, Serena Bonin, Silvia Buriolla, Elisa Bertoli, Maria Antonietta Pizzichetta, Iris Zalaudek, and Fabio Puglisi. 2021. "Find the Flame: Predictive Biomarkers for Immunotherapy in Melanoma" Cancers 13, no. 8: 1819. https://doi.org/10.3390/cancers13081819

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop