Next Article in Journal
Understanding Carbohydrate Metabolism and Insulin Resistance in Acute Intermittent Porphyria
Next Article in Special Issue
MMR Deficiency Defines Distinct Molecular Subtype of Breast Cancer with Histone Proteomic Networks
Previous Article in Journal
Sensorimotor Cortical Activity during Respiratory Arousals in Obstructive Sleep Apnea
Previous Article in Special Issue
Comprehensive Analysis of N6-Methyladenosine (m6A) Writers, Erasers, and Readers in Cervical Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Insights and Strategies of Melanoma Immunotherapy: Predictive Biomarkers of Response and Resistance and Strategies to Improve Response Rates

by
Attila A. Seyhan
1,2,3,4,* and
Claudio Carini
5,6
1
Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
2
Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
3
Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, RI 02912, USA
4
Legorreta Cancer Center, Brown University, Providence, RI 02912, USA
5
School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, New Hunt’s House, Guy’s Campus, King’s College London, London SE1 1UL, UK
6
Biomarkers Consortium, Foundation of the National Institute of Health, Bethesda, MD 20892, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(1), 41; https://doi.org/10.3390/ijms24010041
Submission received: 19 November 2022 / Revised: 10 December 2022 / Accepted: 16 December 2022 / Published: 20 December 2022
(This article belongs to the Special Issue Altered RNA Processing in Tumor Pathogenesis and Therapy)

Abstract

:
Despite the recent successes and durable responses with immune checkpoint inhibitors (ICI), many cancer patients, including those with melanoma, do not derive long-term benefits from ICI therapies. The lack of predictive biomarkers to stratify patients to targeted treatments has been the driver of primary treatment failure and represents an unmet medical need in melanoma and other cancers. Understanding genomic correlations with response and resistance to ICI will enhance cancer patients’ benefits. Building on insights into interplay with the complex tumor microenvironment (TME), the ultimate goal should be assessing how the tumor ’instructs’ the local immune system to create its privileged niche with a focus on genomic reprogramming within the TME. It is hypothesized that this genomic reprogramming determines the response to ICI. Furthermore, emerging genomic signatures of ICI response, including those related to neoantigens, antigen presentation, DNA repair, and oncogenic pathways, are gaining momentum. In addition, emerging data suggest a role for checkpoint regulators, T cell functionality, chromatin modifiers, and copy-number alterations in mediating the selective response to ICI. As such, efforts to contextualize genomic correlations with response into a more insightful understanding of tumor immune biology will help the development of novel biomarkers and therapeutic strategies to overcome ICI resistance.

1. Introduction

Immune checkpoint inhibitors (ICIs) are revolutionizing the treatment of melanoma and other types of cancers, with 7 agents now approved in the US and a rich pipeline of new agents and mechanisms in development [1,2,3,4]. Notwithstanding the excitement around these developments, there is a significant unmet medical need in the form of patient stratification and therapy resistance. Melanoma affects more than 1 million Americans, and there is an increasing incidence of melanoma worldwide. Approx. 300,000 new cases are diagnosed in the US each year [5], with the average annual cost for treatment estimated at $3.3 billion [6].
Numerous successes have been achieved with anti-PD1, anti-CTLA4, or combination therapies [5,7] in treating melanoma and other types of cancers. The groundbreaking finding by Leach et al. [8] showed that antibodies blocking the T cell co-inhibitory receptor CTLA-4 can augment immune responses against tumor cells in mice. This finding gave rise to ipilimumab, the first ICI to increase the survival of patients with metastatic melanoma [9,10] which was granted FDA approval in 2011 for the treatment of metastatic melanoma.
In 2014, additional T cell immune checkpoint blocking antibodies, anti-PD-1 (pembrolizumab) and anti-PD-L1 (nivolumab) [11], received FDA approval. The combination of anti-PD-1 (pembrolizumab) and anti-PD-L1 (nivolumab) resulted in the augmented survival of patients with metastatic melanoma compared to patients treated with ipilimumab alone or chemotherapy alone [12,13,14].
In 2022, relatlimab, which targets lymphocyte-activation gene 3 (LAG-3), was approved by the FDA for adult and pediatric use with metastatic melanoma [15].
Following those developments, high levels of PD-L1 expression in cancer cells and tumor mutational burden (TMB) have been shown in melanoma to correlate with clinical responses to ICI [13].
Despite these data, a substantial number of melanoma patients fail to respond to ICI, leading to premature death [16,17,18]. Considerable effort has been made to identify biomarkers that predict clinical response/resistance to ICI [19]. Despite the successes of ICI, even with the combination of ipilimumab and nivolumab, the five-year survival for the intention-to-treat (ITT) population was only 53%. This means that 47% of patients do not reach long-lasting benefits and succumb to the disease [20], suggesting the need for predictive biomarkers of response to overcome ICI resistance.
Despite the approval of all of these new ICIs and combination therapy, physicians are nowhere near having predictive biomarkers of response to ICI. Presently, physicians have no idea which patients will or will not respond to ICI in the absence of predictive biomarkers of response.
Recently, Carlino et al. [21] demonstrated that combining anti-PD-1 and anti-CTLA-4 ICI in stage IV melanoma resulted in the highest 5-year overall survival rate of all the other therapies. Increased PD-L1 expression and TMB have been shown to correlate well with melanoma response to ICI [13]. Unfortunately, both PD-1 and CTLA4 are unable to predict the outcome [22]. As a result, we run the risk of undertreating some patients who might benefit from ICI.
To date, there are no reliable predictive biomarkers able to stratify responders and non-responders to ICI. Clinical factors, such as volume and disease sites, serum lactate dehydrogenase levels, and BRAF mutation status, used to select the initial therapy for patients with advanced melanoma have been rather unreliable [21].
The clinical trial data of melanoma response to ICI have identified three almost equal populations of patients: (1) those who respond (responders), (2) those who fail to ever respond (innate resistance), and (3) those who initially respond or have a prolonged period of disease stabilization, but eventually develop disease progression (acquired resistance) [7,16,23,24,25].
The current stratification strategies, including genetic mutations and variations in mutational load, have shown some correlation with response to ICI. However, they are unable in predicting patient response to ICI [26]. It has been shown that tumor cells possess genetic and epigenetic traits that facilitate immune evasion [27]. Tumors can mutate to evade the innate and adaptive immune response [19], rendering ICI therapy ineffective [23,28,29]. ICI resistance can start from different cells and their interactions in the tumor ecosystem [29,30]. Recent studies have revealed insights into the mechanism of ICI resistance [7,29,30]. As a result, ICI therapies elicit significantly limited efficacy in many of the cancer types due to drug resistance and toxicity.
The lack of predictive biomarkers to stratify patients to targeted treatments has been the driver of primary treatment failure and represents an unmet medical need in melanoma and other cancers. Therefore, understanding genomic correlations with response and resistance to ICI will be beneficial to cancer patients.
Here, we review recent studies involving ICI therapies in melanoma and other cancers that have revealed important insights about the tumor immune microenvironment in melanoma and other cancers, the mechanisms supporting these findings, and genomic correlations with response and resistance to ICI.
We further discuss how the insights gained from these studies are guiding more precise analyses of the mechanisms of action of ICI therapy and novel immunotherapy approaches, including novel combination therapies to overcome resistance to ICI therapy and turning “cold tumors” into “hot tumors”.
Consequently, these efforts will help us better understand the mechanisms involved in response and resistance to ICI, thus achieving more effective and durable use of immunotherapy.

2. Immune Checkpoint Inhibitors

As illustrated in Figure 1, immunosuppressive mechanisms involving the expression of checkpoint proteins (e.g., PD-1, CTLA-4, LAG-3, TIM-3, and NKG2A/B), activation of cell death programs, and accumulation of various immunosuppressive cells are instrumental in maintaining immune homeostasis and self-tolerance [31,32,33,34]. Cancer cells exploit those immune homeostasis mechanisms to evade the immune system [35]. Several ICI treatments are successful against a wide variety of cancers, given the durability of response and improved side effect profile compared to chemotherapeutic agents. Nonetheless, only a few ICIs have been approved by the FDA [34].

3. Melanoma Biomarkers of Response to ICI

Physicians would greatly benefit in therapeutic decision-making by having predictive biomarker(s) of response to stratify melanoma and other types of cancer patients into responders and non-responders. A general representation of predictive biomarkers of response to ICIs in several cancers is shown in Table 1. However, the significant proportion of non-responders and treatment-associated toxicities and drug resistance remain one of the major obstacles to therapeutic success of ICI in melanoma and other types of cancers.

3.1. Established Clinical Biomarkers

Different types of biomarkers and their clinical utility in metastatic melanoma, including those approved by the FDA and others still at the experimental stage, are shown in Table 2 along with the clinical setting in which they are used.
As summarized in Table 2, the earliest biomarkers that helped to diagnose the presence of melanoma include human melanoma black-45 (HMB-45), melan-A, tyrosinase, microphthalmia transcription factor (MITF), S100, SM5-1, chondroitin sulfate proteoglycan 4 (CSPG4), loss of p16 protein expression, biomarker panels, and gene arrays. Those biomarkers have been used to screen healthy patients before the diagnosis of melanoma in order to help stratify patients into benign vs. malignant stages of the disease.
The earliest approved clinical biomarkers that helped to inform the prognosis of metastatic melanoma relied on baseline clinical characteristics, such as serum levels of lactate dehydrogenase (LDH), a marker of tumor burden [41]. Elevated serum LDH levels have been demonstrated to be a negative prognostic marker, irrespective of the given treatment [42,43,44,45]. Generally, high LDH levels are associated with poor overall survival (OS) compared with normal LDH levels. Elevated levels of LDH have been used as a biomarker to assess patient staging [46]. For example, it has been shown that among ca. 30% of patients with 4–5-year OS following treatment with BRAF and MEK inhibitors, only a few of those patients had high LDH levels before the initiation of therapy [47].
Likewise, elevated serum levels of S100B have demonstrated to have a prognostic value in both metastatic [48,49] and high-risk resected melanoma settings [50].
Gene expression profiling has shown to have a prognostic value that complements existing biomarkers in patients with melanoma [47,51,52].
In metastatic melanoma, a well-characterized predictive biomarker of response guiding the therapeutic decision process is the BRAF V600 mutation, which is somehow predictive of response to BRAF ± MEK inhibition with low rates of primary resistance [53,54,55]. The response rate to BRAF and MEK inhibitors in metastatic melanoma patients with the BRAF V600 mutation is ca. 70% in selected patients, with less than 10% of patients having the highest response to progressive disease [55,56,57,58].
Several oncogenic driver mutations have been identified as predictive biomarkers of response from targeted agents, including NRAS, NF-1, and c-KIT, that provide insight into the probability of therapeutic response to a specific treatment [59,60,61,62,63]. Presently, the presence of a BRAF V600 mutation is the only validated predictive biomarker for melanoma patients.

3.2. Emerging Predictive Biomarkers

As shown in Table 2, many emerging biomarkers are currently being evaluated in clinical studies. Du et al. [64] reported that several genomic and transcriptomic-based biomarkers have been explored as potential predictors of ICI response.
Predictive biomarkers of response include TMB, neoantigen load [7,65,66,67,68,69], HLA-I genotype [70,71], cytolytic activity [72], aneuploidy [73], and T cell repertoire [46], which exhibit high predictability to ICI response. Other predictive biomarkers of response are PDL-1 expression, LAG-3 expression, CD8+ T cells at tumor invasive margin, IL-17 expression, immune-related gene expression signatures, and T-cell receptor (TCR) signature.
Additional biomarkers that are used to assess ICI response include ctDNA profiles, absolute lymphocyte count, proliferating CD8+ T cells, increase of T-cell subsets and checkpoint molecules (PDL-1, LAG-3), granzyme B expression, and T-cell receptor (TCR) signature [37,38,39,40].
Unfortunately, many of those potential biomarkers of ICI response have not yet been validated [74,75,76,77,78].
A recent study by Carter et al. [74] questioned the validity of the immuno-predictive score (IMPRES), a predictor of ICI response in melanoma consisting of 15 pairwise transcriptomic signatures that analyze the relationship between immune checkpoint genes reported by Auslander et al. [76]. The IMPRES is context-dependent and could not reproducibly predict ICI response in the context of metastatic melanoma [76].
Moreover, Xiao et al. [77] questioned the reproducibility of the Immune Cells.Sig [79] signature in melanoma, demonstrating inconsistencies in the prediction capability of ImmuneCells.Sig across different RNA-seq datasets [77]. The performance of the ImmuneCells.Sig signature in predicting ICI outcomes in four melanoma patient datasets, using the same implementation scheme as Xiong et al. [79], showed that there were inconsistencies across different datasets [77].

3.2.1. Gene Expression Signatures

Gene expression signatures (GES) have also been identified as predictive biomarkers of response to ICI and have been validated in several independent datasets (e.g., immune-predictive score, IMPRES consisting of 15 immune genes). IFN-γ-responsive genes were also used to predict ICI response in metastatic melanoma [30,46,72,73,76,80,81,82,83,84,85,86].
A panel of pan-tumor T cell-inflamed GES consisting of 18 IFN-γ-responsive genes was validated and confirmed to predict the response to ICI in pre-treatment tumor specimens from nine types of cancers, including melanoma [83].
MHC-I/II gene signatures have also been explored as predictive biomarkers of ICI response in melanoma [85,87,88].
Carter et al. [74] reported that immuno-predictive score (IMPRES), a predictor of ICI response in melanoma encompassing 15 pairwise transcriptomics relations between immune checkpoint gene [76], did not reproducibly predict the response to ICI in metastatic melanoma.
It was argued that many factors may contribute to the limited successes of those biomarkers, such as: (1) the predictive biomarkers have been derived from pre-clinical studies; (2) evaluation of the biomarkers in clinical specimens only included baseline biopsies and peripheral blood samples; (3) batch effect, lack of reproducibility might have contributed to the failure of the biomarkers for ICI response [74,75,77,78].
To address these issues, numerous researchers have developed predictive biomarkers to reduce batch noise and other technical issues. Expanded predictive biomarker panels have resulted in higher reproducibility as opposed to predictive signatures based on individual biomarkers [89,90,91,92].
Tian et al. [91] reported that the combined BRAF, KRAS, and PI3KCA mutation signature resulted in a favorable predictive response to cetuximab for patients with colorectal cancer [91].

3.2.2. Gene Expression Signatures at Baseline and On-Treatment Tumor Specimens

Genome-wide analysis of transcriptomic and genomic profiles of baseline and on-therapy tumor specimens from patients treated with ICI provides a comprehensive view into the mechanisms underlying tumor response and resistance to ICI [93].
Grasso et al. [93] reported that the mechanism of action of ICI is based on the interaction between immune effector cells and cancer cell targets. Tumor studies conducting comprehensive analyses of transcriptomic and genomic profiling have focused not only on the genetic alterations and gene expression profiles of cancer cells [25,30,85,88], but also on the composition of immune infiltrates and expression of immune-activating gene programs [36,46,67,75,80,82,83,85,86,88,94,95,96,97,98].
Du et al. [64] reported that pathway-based signatures derived from on-treatment tumor specimens were predictive of the response to anti-PD1 blockade in patients with metastatic melanoma.
Other studies of breast cancer suggested that post-treatment tumor samples were more informative than pre-treatment samples [99,100,101].
Conversely, Wallin et al. developed adaptive immune signatures based on tumor samples obtained during the early course of treatment, showing that the signatures were highly predictive of the response to ICI in patients with metastatic melanoma [102].
Auslander et al. built an immune-predictive score (IMPRES), which encompasses 15 pairwise transcriptomic relations between immune checkpoint genes, to predict the response of metastatic melanoma to ICB therapy [76].
The IMPRES signature produced better predictive scores with post-treatment samples than with pre-treatment samples in two independent datasets [76].
In support of these findings, a recent proteome profiling study of samples from patients with metastatic melanoma undergoing either tumor infiltrating lymphocyte based or anti-PD1 immunotherapy demonstrated that the fatty acid oxidation pathway was significantly enriched in responder patients. These results underlined the critical role of mitochondrial metabolism, including fatty acid metabolism, in conferring response to immunotherapy [103].
It is now widely accepted that post-treatment tumor specimens are generally much more informative than pre-treatment specimens and may provide more valuable insight into dynamic changes at the transcriptional level that correlate with clinical response, resulting in a higher predictive score.
In conclusion, although pathway signatures derived from post-treatment samples are highly predictive of therapeutic response to anti-PD1 in patients with metastatic melanoma, further studies are warranted to confirm the predictive value of those signatures in larger cohorts of patients with metastatic melanoma.

3.2.3. Pathway Signatures

Du et al. [64] developed pathway-specific signatures in pre-treatment (PASS-PRE) and on-treatment (PASS-ON) tumor specimens based on transcriptomic data and clinical information from a large dataset of metastatic melanoma patients treated with anti-PD1. Both PASS-PRE and PASS-ON signatures were validated in three independent datasets of metastatic melanoma. Compared to existing molecular signatures, it was concluded that the on-treatment (PASS-ON) tumor specimen signature exhibited a robust and better predictive value for metastatic melanoma patients who responded to anti-PD1 across all four datasets.
The pre-treatment pathway signatures included six pathways for predicting the response to anti-PD1 treatment, including: (1) complement cascade; (2) regulation of insulin-like growth factor IGF transport and uptake by insulin-like growth factor binding proteins IGFBPS; (3) binding and uptake of ligands by scavenger receptors; (4) plasma lipoprotein remodeling; (5) IL2 family signaling; and (6) retinoic acid (RA) biosynthesis pathways. Complement cascade, binding, and uptake of ligands by scavenger receptors and IL2 family signaling pathways are related to immune and inflammation, whereas plasma lipoprotein remodeling and the RA biosynthesis pathways are related to metabolism.
In contrast to the pathway-based signature analysis of on-treatment samples, Du et al. [64] identified four pathways, including: (1) peroxisomal lipid metabolism; (2) generation of second messenger molecules; (3) fatty acid metabolism; and (4) PD1 signaling. Of note, peroxisomal lipid metabolism and fatty acid metabolism are related to fatty acid and lipid metabolism [103]. Generation of second messenger molecules is a central signaling pathway in T-cell receptor (TCR) stimulation. Likewise, PD1 signaling plays an important role in immunoregulation as an immunoregulatory signaling pathway.
To further validate the predictive performance of a pathway-based super signature for on-treatment samples (PASS-ON), Du et al. [64] tested three independent datasets with RNA-seq data available for on-treatment samples, including those of Gide et al. and Lee et al. [96,104], and the MGH cohort demonstrated the effectiveness of PASS-ON in predicting patient response to anti-PD1. Patients with high PASS-ON signature scores were associated with significantly improved PFS compared to those with low signature scores in all tested patients.
Furthermore, Du et al. [64] demonstrated that the time-response interaction pathway-based super signature for pre- and on-treatment samples had reasonable predictive power. The study suggested that pathway-based biomarker signatures derived from on-treatment tumor specimens compared to pretreatment tumor specimens were better predictors of response to anti-PD1 therapies in metastatic melanoma patients.

3.2.4. Tumor Antigens

Huang et al. [22] investigated several melanoma-relevant tumor-specific antigens, cancer germline genes, melanocyte differentiation antigens, overexpressed antigens, neoantigens, neuropeptides, and other sources of immunogenic antigens, such as immunogenic epitopes, have also been explored as novel predictive biomarkers of ICI response to melanoma.
Neoantigens are derived from tumor-specific somatic mutations and are exclusively expressed in cancer cells and absent in normal human tissue. The majority (95%) of somatic mutations are single-nucleotide variants (SNVs), which lead to aberrant protein and peptide expression with single amino acid substitutions [105].
Neopeptides also arise from nucleotide insertions or deletions (indels), leading to the expression of aberrant proteins and peptides with frameshift or non-frameshift sequences depending on the number of nucleotides added and deleted. While the minority of mutations are indels (<5% for melanoma) [106,107], frameshift mutations can generate a number of immunogenic neoepitopes that are highly distinct from the self.
Other sources of immunogenic antigens, including immunogenic epitopes, can also derive from mutations associated with gene fusion, aberrant messenger-RNA splicing with retained introns, or aberrant translation resulting in cryptic antigens, and genomically integrated endogenous retroviral sequences as a result of previous retroviral infections, although they are epigenetically silenced, can be reactivated in tumors [106], as in the case of cancer germline antigens.
Furthermore, tumors often present aberrant patterns of DNA methylation, resulting in the demethylation, ectopic expression, and presentation of cancer germline genes to T cells relevant in immune recognition [106,108].
For example, cancer germline genes such as MAGEA1 and NY-ESO-1 are silenced epigenetically through methylation in human tissue, with the exception of male germ cells and trophoblastic cells, which lack MHC-I molecules.
PRAME (preferentially expressed antigen in melanoma), a member of the cancer-testis antigen family, has been reported to be frequently overexpressed in many cancers, including melanoma, which indicates advanced cancer stages and poor clinical prognosis [109,110]. As such, overexpressed PRAME is a potential immunotherapy target. PRAME-specific immunotherapies are currently in development for many cancers, including melanoma. For example, a recent study demonstrated that uterine carcinosarcoma, synovial sarcoma, and leiomyosarcoma patients would potentially benefit from PRAME-specific immunotherapies [109].

3.2.5. Genomic Alterations

Considerable effort has been made to identify genomic alterations and transcriptome profiles as predictive biomarkers of ICI response. Numerous studies have identified distinct stages of CD8+ T cells linked to positive response or failure to ICI treatment [98].
Moreover, tumors from patients responding to ICI showed a higher number of cancer-associated somatic mutations (i.e., mutated antigens or neoantigens) targeted by T cells [65]. The IFN-γ signature has also been shown to predict the response to ICI (e.g., anti-PD-1) in melanoma [111] and in other types of cancers [6,93]. Gene expression signatures obtained from bulk melanoma tumor or single-cell profiling and the TME have been shown to be correlated with sensitivity and resistance to several ICIs [86,88,98,112,113,114].
Collectively, the gene expression signatures associated with response to ICI in metastatic melanoma represent distinct characteristics and play an important function in different signaling pathways, including the inflammatory response, type I interferon signaling pathway, cytokines, and others.

4. Who Is Responding to ICI?

Recent data have demonstrated that immunotherapies against immune checkpoints (e.g., CTLA-4 or PD-1) downregulate two main negative regulators of the anti-tumor immune response [93,115,116,117], resulting in durable anti-tumor responses in a subset of cancer patients, including those with melanoma [2,118].
Another key factor contributing to anti-tumor immune response following ICI treatment [93] is the pre-existing level of T cell infiltration of the tumor [119,120,121], representing the immunogenicity of the cancer cells.
Analysis of tumor biopsies from ICI-treated patients showed that clinical responses associated with ICI were mediated by tumor-infiltrating T cells reactivated following ICI treatment [121,122].
A combination of immunohistochemical (IHC) analysis with RNA-seq performed on cancer biopsies from patients treated with anti-CTLA-4 antibody (ipilimumab) before or after treatment with anti-PD-1 antibody (nivolumab) demonstrated that a major response to anti-CTLA-4 requires cancer cells with high levels of MHC-I expression at baseline, whereas the response to anti-PD-1 was more strongly associated with a pre-existing interferon-γ gene expression signature [88].
Liu et al. demonstrated that the response to anti-PD-1 therapy (with or without prior anti-CTLA-4 treatment) was associated with increased MHC-I and MHC-II expression [85]. This study demonstrated that patients not responding to therapy have occasional genetic alterations in antigen presentation genes [85].
Biopsies from patients with metastatic melanoma treated with anti-PD-1 monotherapy (nivolumab) in part 1 of the CheckMate 038 study showed an increase in immune cell subtypes with elevated immune activation gene signatures seen in responders to therapy [25].
The transcriptome analysis of tumor biopsies from patients treated with anti-PD-1 monotherapy (nivolumab) or in combination anti-PD1 plus anti-CTLA-4 therapy (ipilimumab) correlated well with the in vitro analysis of gene expression signatures of melanoma cell lines following exposure to interferon-γ [93].
It appears that cancer cells become enablers of the immune response via the expression of IFN-γ-response genes, triggering the upregulation of antigen presentation, amplification of the interferon response, and induction of chemokines (i.e., CXCL9 and CXCL10) to entice immune cells to the TME. Thus, T cell-induced IFN-γ correlates with ICI therapy response [93]. Collectively, the degree of the anti-tumor T cell response and downstream IFN-γ signaling are the main drivers of response or resistance to ICI therapy [93].
However, little is known about how tumor-intrinsic loss of IFN-γ signaling impacts TILs. The question remains whether tumor-intrinsic IFN-γ signaling actively regulates the infiltration or function of TILs?
Shen et al. [123] demonstrated that IFN-γR1 knockout melanomas and IFN-γR1KO melanomas in B6 mice had reduced infiltration and function of tumor-infiltrating lymphocytes (TILs). Furthermore, long-distance effects of IFN-γ on tumor cells also play a crucial role in anti-tumor immunity [123]. These recent findings revealed an important role of tumor-intrinsic IFN-γ signaling and IFN-γ-response genes in shaping TILs.

5. Tumor-Immunity Cycle and Resistance Mechanisms Involving Tumor Immunophenotypes

As discussed in the literature [124] and illustrated in Figure 2 and Figure 3, the anti-tumor-immunity cycle is a gradual process mediated to a large extent by CD8+ T lymphocytes and involves a multi-step process.
In the immune-desert phenotype, immune cells are absent from the tumor and its periphery. In the immune-excluded phenotype, immune cells accumulate but do not efficiently infiltrate. In the immune-inflamed phenotype, immune cells infiltrate but their effects are inhibited. Notably, the three different phenotypes have different response rates to immune checkpoint inhibitors.
Based on the spatial distribution of cytotoxic immune cells in the TME, a tumor is classified as an immune-inflamed, immune-excluded, or immune-desert phenotype (Figure 2 and Figure 3) [126]. Immune-inflamed tumors (i.e., “hot tumors”) are characterized by increased T cell infiltration, high interferon-γ (IFN-γ) signaling, elevated expression of PD-L1, and increased TMB [127]. Inflamed tumors are more responsive to ICIs than non-inflamed tumors [119,128]. Immune-deserted tumors (i.e., “cold tumors”), on the other hand, exhibit characteristics where CD8+ T lymphocytes localize only at the tumor margin and do not infiltrate the tumor [129]. In immune-desert tumors, CD8+ T lymphocytes are absent from the actual tumor and its periphery [129]. In addition to poor T cell infiltration, “cold tumors” display low mutational load, decreased MHC class I expression, and reduced PD-L1 expression [127].
Cold tumors also harbor immunosuppressive cells, including T-regulatory cells (Tregs) and myeloid-derived suppressor cells (MDSCs), and tumor-associated macrophages (TAMs) are key sources of many of these inhibitory factors [127]. As a result, cold tumors lack innate immunity or the innate antitumor immune features present in cold tumors’ may be ineffective due to the lack of immune cells [126]. The three tumor immune phenotypes have different response rates to ICIs and cold tumors respond poorly to ICI monotherapy [119].

5.1. Tumor Cell-Intrinsic and Tumor Cell-Extrinsic Resistance Mechanisms

Many factors play a role in T cells driving tumor resistance, ultimately leading to a noninflamed T cell phenotype and failed antitumor immunity (Figure 3 and Figure 4).
Several other resistance mechanisms involving both tumor cell-intrinsic and tumor cell-extrinsic sources have been described (Figure 3 and Figure 4 and Table 3) [34]. In the case of tumor cell-intrinsic mechanisms, a lack of neoantigen development, impaired antigen presentation, and other primary factors contribute to the resistance to immunotherapy [130,131,132,133,134,135,136]. Tumor cell-extrinsic mechanisms encompass increased recruitment and activity of inhibitory immune cells within the TME and upregulation of LAG-3 and TIM-3 [16,47,137,138,139,140].

5.2. Immune Resistance Mechanisms in Melanoma

As illustrated in Figure 2, Figure 3, Figure 4 and Figure 5 and Table 3, resistance to ICI therapy is one of the most significant challenges to achieving a durable tumor response in many types of cancers [16,24,34,182,183].
Several mechanisms, including primary (lack of tumor response to initial immunotherapy) and secondary/acquired resistance to cancer immunotherapy (initial response to tumor followed by a lack of response) have been described as the major resistance mechanisms to ICI therapy [16] (Figure 2, Figure 3, Figure 4 and Figure 5 and Table 3).
As for melanoma, Huang et al. [22] demonstrated that several key mechanisms are involved in the immune resistance to ICI, such as immune tolerance, T cell exhaustion, immune cell-mediated immunosuppression, expression of immune checkpoint ligands, and tumor escape.
Immune tolerance describes the lack of response by the immune system to substances or antigens that have the potential to induce an immune response. Immune tolerance to an individual’s own antigens occurs through both central and peripheral tolerance mechanisms. While central tolerance occurs via thymic deletion of high-affinity auto-reactive T cells, peripheral tolerance is maintained by other mechanisms (e.g., suppression by Treg cells and anergy) and induced by many mechanisms, including sub-optimal T cell co-stimulation, deletion via apoptosis, or conversion into Treg cells. The dose of antigen and TCR affinity are considered to be the major drivers of these mechanisms [22]. Notably, both the lymph node and tumor environments blunt T-cell effector functions and offer a rationale for the failure of tumor-specific responses to effectively counter tumor progression [186].
T cell exhaustion is a specific T cell differentiation process mediated by chronic antigen stimulation, which leads to increased expression of co-inhibitory immune receptors that are presumed to decrease chronic TCR signaling and regulate activation-induced cell death.
In this state of simultaneous TCR stimulation and co-inhibitory pathway stimulation, exhausted T cells (TEX cells) exhibit reduced effector functions (i.e., cytokine production and proliferative potential), but can survive in the hostile TME. Notably, T cell exhaustion appears to be a dynamic and progressive process that includes intermediate reversible states more permissive to stimulation by ICI.
Cell-mediated immunosuppression involves immunosuppressive cells, including MDSCs, Treg cells, and tolerogenic DCs, instructing effector T and B cells to not respond to positive immune stimuli.
Upregulation of immune checkpoint ligands PD-L1 and PD-L2 is often seen in melanoma and other cancers in response to robust inflammatory signals as a homeostatic mechanism adopted by cancer cells to shield themselves from immune attack. Interaction of PD-L1 and PD-L2 ligands through binding to PD-1 receptors expressed on tumor-specific T cells initiate a negative signaling process downstream of PD-1, which reduces T cell activation and impairs tumor-killing function [11]. As a consequence, expression of PD-L1 or PD-L2 in melanoma cells can neutralize the positive T cell signals mediated by the MHC-I and MHC-II antigen presentation process.
Tumor escape is a mechanism that cancer cells utilize to escape from anti-tumor immunity and immune surveillance. Tumor escape can be mediated by tumor-extrinsic mechanisms in the TME and by the tumor itself, which can evolve to evade immune recognition. Under strong immune selective pressure, heterogeneous tumor cells can result in clonal evolution, selection, and enrichment of specific tumor cells that can evade immune recognition, leading to immunotherapy resistance. The evasion of immune recognition by these treatment-resistant cells can take place via inactivation of antigen-presentation processes (e.g., B2M, HLA, TAP, etc.) and/or IFN-γ-response genes (e.g., JAK1, JAK2).

6. Therapeutic Strategies to Turn “Cold Tumors” into “Hot Tumors”

Several strategies have been investigated to elucidate the mechanisms underlying how T cells are driven into “hot tumors” in order to improve the efficacy of ICI therapy (Figure 6 and Table 4) [124]. Several clinical trials have tested these novel therapeutic modalities as interventions in combination with ICI to overcome ICI monotherapy resistance and attempt to turn “cold tumors’ into “hot tumors” (Table 5).
Grasso et al. [93] showed that a robust anti-tumor immune response relies on the interplay of key factors that can be modulated with innovative interventions.
Recent preclinical and clinical findings have provided insight into the immunological implications of canonical cancer signaling pathways (e.g., WNT-beta-catenin signaling, cell cycle regulatory signaling, mitogen-activated protein kinase signaling, and pathways activated by loss of the tumor suppressor phosphoinositide phosphatase PTEN), thus providing new opportunities for the development of new treatments for those patients who do not respond to ICI monotherapies [218].
Combined therapeutic strategies from preclinical [115,117,219] and clinical studies [14] have shown that anti-PD-1 plus anti-CTLA-4 (nivolumab and ipilimumab) treatments elicit stronger immune stimulation in stage IV melanoma than monotherapy alone, resulting in a favorable anti-tumor immune response. Response to ICI, either following anti-CTLA-4 monotherapy or in combination with anti-PD-1 therapy, triggers a robust T cell response that generated an appreciable antitumor response [218].
Another approach to turn “cold tumors” into “hot tumors” is through the intra-tumoral delivery of oncolytic viruses or Toll-like receptor agonists capable of inducing intra-tumoral interferon production, which triggers the pattern recognition pathways with consequent boosting of the anti-PD1 immune response rate [187,190,220].
Recently, different approaches have been used to boost the response to ICI. Activation of the STING pathway [221], inhibition of immune suppressive factors (e.g., WNT signaling or the adenosine pathway) [111,209,222,223], as well as the release of other immune checkpoints (e.g., LAG-3, TIM-3, or TIGIT, etc.) in T cells [111] have been explored, but no favorable clinical outcomes have yet been reported in patients.
Altogether, these new therapeutic strategies provide new opportunities for cancer immunotherapy for patients who do not respond to ICI [111].

7. Lessons Learned: ICI Therapies in Melanoma

Huang et al. [22] reported that several factors, including the immune TME, tumor-associated immune cells, and different host factors, contribute to the ICI resistance. Melanoma resistance has armed us with a handful of information that can be applied to other types of cancers, such as: (1) the ability to present cancer antigens through MHC-I and elevated TMB; (2) tumor-antigen-specific T cells play a crucial role in the response to ICI; (3) reactivation of terminally exhausted T cells could be considered a biomarker for PD-1 blockade, which is detectable as early as one week after ICI dosing; (4) melanoma immunosuppressive mechanisms are complicated and need additional research to remodel their interaction, cooperation, and dynamics during tumor progression and in immunotherapy resistance; and (5) Treg cells are emerging as a key mechanism of resistance to PD-1 blockade, but not necessarily CTLA-4 blockade; (6) emerging neoadjuvant immunotherapy trials are anticipated to provide new insight into pharmacodynamic immune responses and advance the development of rational immunotherapy and neoadjuvant combination regimens while avoiding toxicity and significantly improving patient management; (7) longitudinal assessment of pre- and on-treatment patient specimens is required to determine prognostic vs. predictive use of immune and other parameters, including genomic parameters correlating with patient outcomes, and deduce their biologic role in response to ICI therapy based on their modulation during treatment; and last but not least (8) melanoma-specific oncogenic programs supporting metabolic plasticity and fitness, together with clinical and preclinical evidence of differential activity of ICI therapy depending on the tumor metabolic state, should provide new research opportunities to evaluate these relationships as potential biomarkers for patient stratification and treatment allocation, and formulate novel precision-medicine combinations depending on metabolic and immune therapies.

8. Conclusions and Future Perspectives

Even with a high level of immunogenicity, metastatic melanoma grows and spreads rapidly via escape mutations and immunosuppression. Although combination ICI therapies targeting CTLA-4 and PD-1 can efficiently target some ICI-resistant mechanisms by improving T cell priming, re-activating PD-1 high CD8 + exhausted T cells, and reversing Treg suppression, many patients still do not derive durable clinical benefit.
Therefore, with the availability of novel immunotherapeutic agents, the mechanism of single-agent therapies needs to be better characterized in order to guide effective rational ICI combinations. In addition, since the immunologic effects of ICI therapy occur early, we need to focus on these early events to identify specific biomarkers, mechanisms of resistance, and neoadjuvant therapies.
Furthermore, toxicity from current and new ICI combinations remains a critical step to be addressed [224]. Because of concerns regarding adverse events from current and new ICI combinations, insight gained from molecular mediators of immune toxicity (e.g., antibody- vs. T cell-mediated) resulting from combination ICI therapies would help in avoiding these side effects and the development of novel combination ICI therapies to other cancers.
As a result, new efficacy and toxicity data deriving from different ICI therapies across various tumor types will help to characterize key parameters for predicting ICI response, thus limiting toxicity and enhancing therapeutic decision processes to overcome ICI resistance. In fact, new efficacy and toxicity data from many ICI therapies across various tumor types are helping in the characterization of key parameters for predicting response, limiting toxicity, and informing therapeutic decisions to overcome resistance.
Furthermore, the identification of novel genomic correlations with resistance to ICI requires well-annotated data from diverse patient cohorts and tumor histology to detect rare response-associated variants [159,225]. With a greater focus on detailed genomics as well as epigenomics, proteomics, and metabolomics, microbiome and biomarker studies will better characterize the relationships among host immunity, tumor biology, and mechanisms of resistance and response to ICI. Ultimately, molecular and clinical data from these studies will have to be adequately integrated and blended into preclinical studies.
For this reason, systems biology, computational biology, biostatistics, as well as machine learning and artificial intelligence approaches must be coordinated to integrate and help interpret this large dataset, thus translating the findings into actionable novel biomarkers that capture the complexity of multiple alterations affecting response and therapeutic success.
In addition, several other key challenges face ICI therapy that must be addressed to move the field ahead [226], including: (1) development of preclinical models that translate to human immunity; (2) identification and validation of the dominant drivers involved in cancer immunity; (3) deeper insights into organ-specific immune TME; (4) exploration of differences in molecular and cellular drivers between primary and secondary immune escape; (5) characterization of the benefit of endogenous vs. synthetic immunity; (6) investigation of ICI therapy combinations with other drugs (Figure 6, Table 4 and Table 5) in early-phase clinical studies; (7) investigation of steroids and immune suppression on immunotherapy and autoimmune adverse events and toxicities; (8) boosting personalized medicine approaches by using composite biomarkers; (9) developing and implementing refined regulatory endpoints for immunotherapy; and (10) optimizing durable survival with a combination of multi-agent immunotherapy regimens [226].
Consequently, all of these efforts will result in a better understanding of the mechanisms involved in response and resistance to ICI, hence facilitating the development of biomarkers and novel therapies.
In conclusion, these efforts should result in a deeper quantitative and conceptual understanding of the mechanisms involved in ICI response and resistance, thus facilitating the development of rational biomarkers and therapies.

Author Contributions

Conceptualization, A.A.S. and C.C.; methodology, A.A.S. and C.C.; software, A.A.S.; validation, A.A.S. and C.C.; formal analysis, A.A.S. and C.C.; investigation, A.A.S. and C.C.; resources, A.A.S. and C.C.; data curation, A.A.S.; writing—original draft preparation, A.A.S.; writing—review and editing, A.A.S. and C.C.; visualization, A.A.S.; supervision, A.A.S. and C.C.; project administration, A.A.S. and C.C. All authors have read and agreed to the published version of the manuscript.

Funding

No research funding received from specific grant from any funding agency in the public, commercial, or not-for-profit sectors was used in the preparation of this review article.

Institutional Review Board Statement

Not applicable.

Data Availability Statement

The authors grant the publisher the sole and exclusive license of the full copyright in the Contribution, which license the publisher hereby accepts.

Acknowledgments

We thank the Legorreta Cancer Center at Brown University for providing the environment for the preparation of this review article. The contents of this manuscript are solely the responsibility of the authors.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

A2aRadenosine A2a receptor
ADOadenosine
AEadverse event
APCantigen-presenting cell
ATPadenosine triphosphate
B2Mbeta-2-microglobulin
B7RP1B7-related protein 1
BATF3basic leucine zipper ATF-like transcription factor 3
BTLAB and T lymphocyte attenuator
CAFcancer-associated fibroblast
CCR4C-C chemokine receptor type 4
CD40LCD40 ligand
CDNcyclic dinucleotide
CRTCalreticulin
CSF-1colony stimulating factor 1
CTCscirculating tumor cells
ctDNAcirculating tumor DNA
CTLA-4cytotoxic T-lymphocyte-associated protein 4
CXCL/CCLchemokine motif ligands;
dMMRdefective mismatch repair
DNMTDNA methyltransferase
ECMextracellular matrix
EGFRepidermal growth factor receptor
ERendoplasmic reticulum
ETBRendothelin B receptor
EZH2enhancer of zeste homolog 2
FDAFood and Drug Administration
FLT3LFms-like tyrosine kinase 3 ligand
GAL9galectin 9
GITRglucocorticoid-induced tumor necrosis factor receptor
GM-CSFgranulocyte-macrophage colony-stimulating factor
HDAChistone deacetylase
HER2human epidermal growth factor receptor 2
HEVhigh endothelial venule
HLAhuman leukocyte antigen
HMGB1high mobility family protein B1
HRhormone receptor positive
HVEMherpesvirus entry mediator
ICAMintercellular adhesion molecule
ICOSinducible T cell co-stimulator
IDOindoleamine 2,3-dioxygenase
IFNinterferon
ILinterleukin
ITTintention-to-treat
KIRkiller cell immunoglobulin-like receptors
LAG-3lymphocyte activation gene 3
LFA1lymphocyte function-associated antigen-1
mAbmonoclonal antibody
M-CSFmacrophage colony-stimulating factor
MDSCmyeloid-derived suppressor cell
MHCmajor histocompatibility complex
MMRmismatch repair
MSImicrosatellite instability
MSI-Hmicrosatellite instability-high
MɸIItype II macrophage
NKnatural killer
OSoverall survival
OX40LOX40 ligand
PD-1programmed cell death protein-1
PD-L1programmed death-ligand 1
PD-L2programmed death-ligand 2
PFSprogression-free survival
PI3Kphosphatidyl-inositol 3-kinase
PTENphosphatase and tensin homolog
STC1stanniocalcin 1
TAMtumor-associated macrophage
TAPtransporter associated with antigen processing
TCRT-cell receptor
TGFtransforming growth factor
TGFβtransforming growth factor-beta
TILstumor-infiltrating lymphocytes
TIM-3T cell immunoglobulin and mucin-domain containing-3
TLRToll-like receptor
TLStertiary lymphoid structure
TMBtumor mutation burden
TMEtumor microenvironment
TNFtumor necrosis factor
TNFRSF14 tumor necrosis factor receptor superfamily member 14
TregT-regulatory cell
VCAMvascular cell adhesion molecule
VEGFvascular endothelial growth factor
VISTAV-domain Ig suppressor of T cell activation

References

  1. Sharma, P.; Wagner, K.; Wolchok, J.D.; Allison, J.P. Novel cancer immunotherapy agents with survival benefit: Recent successes and next steps. Nat. Rev. Cancer 2011, 11, 805–812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Sharma, P.; Allison, J.P. Immune checkpoint targeting in cancer therapy: Toward combination strategies with curative potential. Cell 2015, 161, 205–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Darvin, P.; Toor, S.M.; Sasidharan Nair, V.; Elkord, E. Immune checkpoint inhibitors: Recent progress and potential biomarkers. Exp. Mol. Med. 2018, 50, 1–11. [Google Scholar] [CrossRef] [Green Version]
  5. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2018. CA Cancer J. Clin. 2018, 68, 7–30. [Google Scholar] [CrossRef] [PubMed]
  6. Ayers, M.; Lunceford, J.; Nebozhyn, M.; Murphy, E.; Loboda, A.; Albright, A.; Cheng, J.; Kang, S.P.; Ebbinghaus, S.; Yearley, J.; et al. Relationship between immune gene signatures and clinical response to PD-1 blockade with pembrolizumab (MK-3475) in patients with advanced solid tumors. J. ImmunoTherapy Cancer 2015, 3, P80. [Google Scholar] [CrossRef] [Green Version]
  7. Van Allen, E.M.; 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. Leach, D.R.; Krummel, M.F.; Allison, J.P. Enhancement of antitumor immunity by CTLA-4 blockade. Science 1996, 271, 1734–1736. [Google Scholar] [CrossRef] [Green Version]
  9. Hodi, F.S.; O’Day, S.J.; McDermott, D.F.; Weber, R.W.; Sosman, J.A.; Haanen, J.B.; Gonzalez, R.; Robert, C.; Schadendorf, D.; Hassel, J.C.; et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 2010, 363, 711–723. [Google Scholar] [CrossRef]
  10. Robert, C.; Thomas, L.; Bondarenko, I.; O’Day, S.; Weber, J.; Garbe, C.; Lebbe, C.; Baurain, J.F.; Testori, A.; Grob, J.J.; et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N. Engl. J. Med. 2011, 364, 2517–2526. [Google Scholar] [CrossRef]
  11. Pauken, K.E.; Torchia, J.A.; Chaudhri, A.; Sharpe, A.H.; Freeman, G.J. Emerging concepts in PD-1 checkpoint biology. Semin. Immunol. 2021, 52, 101480. [Google Scholar] [CrossRef]
  12. Schumacher, T.N.; Schreiber, R.D. Neoantigens in cancer immunotherapy. Science 2015, 348, 69–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Zappasodi, R.; Wolchok, J.D.; Merghoub, T. Correction to: Strategies for Predicting Response to Checkpoint Inhibitors. Curr. Hematol. Malig. Rep. 2019, 14, 62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. 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] [Green Version]
  15. Paik, J. Nivolumab Plus Relatlimab: First Approval. Drugs 2022, 82, 925–931. [Google Scholar] [CrossRef] [PubMed]
  16. Sharma, P.; Hu-Lieskovan, S.; Wargo, J.A.; Ribas, A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell 2017, 168, 707–723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Prieto, P.A.; Yang, J.C.; Sherry, R.M.; Hughes, M.S.; Kammula, U.S.; White, D.E.; Levy, C.L.; Rosenberg, S.A.; Phan, G.Q. CTLA-4 blockade with ipilimumab: Long-term follow-up of 177 patients with metastatic melanoma. Clin. Cancer Res. 2012, 18, 2039–2047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Di Giacomo, A.M.; Calabro, L.; Danielli, R.; Fonsatti, E.; Bertocci, E.; Pesce, I.; Fazio, C.; Cutaia, O.; Giannarelli, D.; Miracco, C.; et al. Long-term survival and immunological parameters in metastatic melanoma patients who responded to ipilimumab 10 mg/kg within an expanded access programme. Cancer Immunol. Immunother. 2013, 62, 1021–1028. [Google Scholar] [CrossRef] [PubMed]
  19. 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]
  20. Wolchok, J.D.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Long-Term Outcomes With Nivolumab Plus Ipilimumab or Nivolumab Alone Versus Ipilimumab in Patients With Advanced Melanoma. J. Clin. Oncol. 2022, 40, 127–137. [Google Scholar] [CrossRef]
  21. Carlino, M.S.; Larkin, J.; Long, G.V. Immune checkpoint inhibitors in melanoma. Lancet 2021, 398, 1002–1014. [Google Scholar] [CrossRef] [PubMed]
  22. Huang, A.C.; Zappasodi, R. A decade of checkpoint blockade immunotherapy in melanoma: Understanding the molecular basis for immune sensitivity and resistance. Nat. Immunol. 2022, 23, 660–670. [Google Scholar] [CrossRef] [PubMed]
  23. Gajewski, T.F.; Schreiber, H.; Fu, Y.X. Innate and adaptive immune cells in the tumor microenvironment. Nat. Immunol. 2013, 14, 1014–1022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Pitt, J.M.; Vétizou, M.; Daillère, R.; Roberti, M.P.; Yamazaki, T.; Routy, B.; Lepage, P.; Boneca, I.G.; Chamaillard, M.; Kroemer, G.; et al. Resistance Mechanisms to Immune-Checkpoint Blockade in Cancer: Tumor-Intrinsic and -Extrinsic Factors. Immunity 2016, 44, 1255–1269. [Google Scholar] [CrossRef] [Green Version]
  25. Riaz, N.; Havel, J.J.; Makarov, V.; Desrichard, A.; Urba, W.J.; Sims, J.S.; Hodi, F.S.; Martin-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]
  26. Zhang, N.; Song, J.; Liu, Y.; Liu, M.; Zhang, L.; Sheng, D.; Deng, L.; Yi, H.; Wu, M.; Zheng, Y.; et al. Photothermal therapy mediated by phase-transformation nanoparticles facilitates delivery of anti-PD1 antibody and synergizes with antitumor immunotherapy for melanoma. J. Control. Release 2019, 306, 15–28. [Google Scholar] [CrossRef]
  27. Khong, H.T.; Restifo, N.P. Natural selection of tumor variants in the generation of "tumor escape" phenotypes. Nat. Immunol. 2002, 3, 999–1005. [Google Scholar] [CrossRef] [Green Version]
  28. Pitt, J.M.; Marabelle, A.; Eggermont, A.; Soria, J.C.; Kroemer, G.; Zitvogel, L. Targeting the tumor microenvironment: Removing obstruction to anticancer immune responses and immunotherapy. Ann. Oncol. 2016, 27, 1482–1492. [Google Scholar] [CrossRef]
  29. Restifo, N.P.; Smyth, M.J.; Snyder, A. Acquired resistance to immunotherapy and future challenges. Nat. Rev. Cancer 2016, 16, 121–126. [Google Scholar] [CrossRef]
  30. 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]
  31. Iliopoulos, D.; Kavousanaki, M.; Ioannou, M.; Boumpas, D.; Verginis, P. The negative costimulatory molecule PD-1 modulates the balance between immunity and tolerance via miR-21. Eur. J. Immunol. 2011, 41, 1754–1763. [Google Scholar] [CrossRef] [PubMed]
  32. Han, Y.; Liu, D.; Li, L. PD-1/PD-L1 pathway: Current researches in cancer. Am. J. Cancer Res. 2020, 10, 727–742. [Google Scholar] [PubMed]
  33. Carlsen, L.; Huntington, K.E.; El-Deiry, W.S. Immunotherapy for Colorectal Cancer: Mechanisms and Predictive Biomarkers. Cancers 2022, 14, 1028. [Google Scholar] [CrossRef] [PubMed]
  34. Gravbrot, N.; Gilbert-Gard, K.; Mehta, P.; Ghotmi, Y.; Banerjee, M.; Mazis, C.; Sundararajan, S. Therapeutic Monoclonal Antibodies Targeting Immune Checkpoints for the Treatment of Solid Tumors. Antibodies 2019, 8, 51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Fan, J.; Shang, D.; Han, B.; Song, J.; Chen, H.; Yang, J.M. Adoptive Cell Transfer: Is it a Promising Immunotherapy for Colorectal Cancer? Theranostics 2018, 8, 5784–5800. [Google Scholar] [CrossRef] [PubMed]
  36. Helmink, B.A.; Reddy, S.M.; Gao, J.; Zhang, S.; Basar, R.; Thakur, R.; Yizhak, K.; Sade-Feldman, M.; Blando, J.; Han, G.; et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature 2020, 577, 549–555. [Google Scholar] [CrossRef]
  37. Tarhini, A.; Kudchadkar, R.R. Predictive and on-treatment monitoring biomarkers in advanced melanoma: Moving toward personalized medicine. Cancer Treat Rev. 2018, 71, 8–18. [Google Scholar] [CrossRef] [Green Version]
  38. Weinstein, D.; Leininger, J.; Hamby, C.; Safai, B. Diagnostic and prognostic biomarkers in melanoma. J. Clin. Aesthet. Dermatol. 2014, 7, 13–24. [Google Scholar]
  39. Louie, A.D.; Huntington, K.; Carlsen, L.; Zhou, L.; El-Deiry, W.S. Integrating Molecular Biomarker Inputs Into Development and Use of Clinical Cancer Therapeutics. Front. Pharmacol. 2021, 12, 747194. [Google Scholar] [CrossRef]
  40. Deacon, D.C.; Smith, E.A.; Judson-Torres, R.L. Molecular Biomarkers for Melanoma Screening, Diagnosis and Prognosis: Current State and Future Prospects. Front. Med. (Lausanne) 2021, 8, 642380. [Google Scholar] [CrossRef]
  41. Franzke, A.; Probst-Kepper, M.; Buer, J.; Duensing, S.; Hoffmann, R.; Wittke, F.; Volkenandt, M.; Ganser, A.; Atzpodien, J. Elevated pretreatment serum levels of soluble vascular cell adhesion molecule 1 and lactate dehydrogenase as predictors of survival in cutaneous metastatic malignant melanoma. Br. J. Cancer 1998, 78, 40–45. [Google Scholar] [CrossRef] [PubMed]
  42. Long, G.V.; Eroglu, Z.; Infante, J.; Patel, S.; Daud, A.; Johnson, D.B.; Gonzalez, R.; Kefford, R.; Hamid, O.; Schuchter, L.; et al. Long-Term Outcomes in Patients With BRAF V600-Mutant Metastatic Melanoma Who Received Dabrafenib Combined With Trametinib. J. Clin. Oncol. 2018, 36, 667–673. [Google Scholar] [CrossRef] [PubMed]
  43. Long, G.V.; Grob, J.J.; Nathan, P.; Ribas, A.; Robert, C.; Schadendorf, D.; Lane, S.R.; Mak, C.; Legenne, P.; Flaherty, K.T.; et al. Factors predictive of response, disease progression, and overall survival after dabrafenib and trametinib combination treatment: A pooled analysis of individual patient data from randomised trials. Lancet Oncol. 2016, 17, 1743–1754. [Google Scholar] [CrossRef]
  44. Larkin, J.M.G.; McArthur, G.A.; Ribas, A.; Ascierto, P.A.; Gallo, J.D.; Rooney, I.A.; Chang, I.; Dréno, B. Clinical predictors of response for coBRIM: A phase III study of cobimetinib (C) in combination with vemurafenib (V) in advanced BRAF-mutated melanoma (MM). J. Clin. Oncol. 2016, 34, 9528. [Google Scholar] [CrossRef]
  45. Long, G.V.; Blank, C.; Ribas, A.; Mortier, L.; Carlino, M.S.; Lotem, M.; Lorigan, P.; Neyns, B.; Petrella, T.M.; Puzanov, I.; et al. 1141 Impact of baseline serum lactate dehydrogenase concentration on the efficacy of pembrolizumab and ipilimumab in patients with advanced melanoma: Data from KEYNOTE-006. Eur. J. Cancer 2017, 72, s122–s123. [Google Scholar] [CrossRef]
  46. 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, eaah3560. [Google Scholar] [CrossRef] [Green Version]
  47. Zager, J.S.; Gastman, B.R.; Leachman, S.; Gonzalez, R.C.; Fleming, M.D.; Ferris, L.K.; Ho, J.; Miller, A.R.; Cook, R.W.; Covington, K.R.; et al. Performance of a prognostic 31-gene expression profile in an independent cohort of 523 cutaneous melanoma patients. BMC Cancer 2018, 18, 130. [Google Scholar] [CrossRef]
  48. Hauschild, A.; Michaelsen, J.; Brenner, W.; Glaser, R.; Henze, E.; Christophers, E. Prognostic significance of serum S100B detection compared with routine blood parameters in advanced metastatic melanoma patients. Melanoma Res. 1999, 9, 155–161. [Google Scholar] [CrossRef]
  49. Hauschild, A.; Engel, G.; Brenner, W.; Glaser, R.; Monig, 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]
  50. Tarhini, A.A.; Stuckert, J.; Lee, S.; Sander, C.; Kirkwood, J.M. Prognostic significance of serum S100B protein in high-risk surgically resected melanoma patients participating in Intergroup Trial ECOG 1694. J. Clin. Oncol. 2009, 27, 38–44. [Google Scholar] [CrossRef] [Green Version]
  51. Gerami, P.; Cook, R.W.; Russell, M.C.; Wilkinson, J.; Amaria, R.N.; Gonzalez, R.; Lyle, S.; Jackson, G.L.; Greisinger, A.J.; Johnson, C.E.; et al. Gene expression profiling for molecular staging of cutaneous melanoma in patients undergoing sentinel lymph node biopsy. J. Am. Acad. Dermatol. 2015, 72, 780–785.e3. [Google Scholar] [CrossRef] [PubMed]
  52. Gerami, P.; Cook, R.W.; Wilkinson, J.; Russell, M.C.; Dhillon, N.; Amaria, R.N.; Gonzalez, R.; Lyle, S.; Johnson, C.E.; Oelschlager, K.M.; et al. Development of a prognostic genetic signature to predict the metastatic risk associated with cutaneous melanoma. Clin. Cancer Res. 2015, 21, 175–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Chapman, P.B.; Hauschild, A.; Robert, C.; Haanen, J.B.; Ascierto, P.; Larkin, J.; Dummer, R.; Garbe, C.; Testori, A.; Maio, M.; et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N. Engl. J. Med. 2011, 364, 2507–2516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Hauschild, A.; Grob, J.J.; Demidov, L.V.; Jouary, T.; Gutzmer, R.; Millward, M.; Rutkowski, P.; Blank, C.U.; Miller, W.H., Jr.; Kaempgen, E.; et al. Dabrafenib in BRAF-mutated metastatic melanoma: A multicentre, open-label, phase 3 randomised controlled trial. Lancet 2012, 380, 358–365. [Google Scholar] [CrossRef] [PubMed]
  55. Long, G.V.; Stroyakovskiy, D.; Gogas, H.; Levchenko, E.; de Braud, F.; Larkin, J.; Garbe, C.; Jouary, T.; Hauschild, A.; Grob, J.J.; et al. Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: A multicentre, double-blind, phase 3 randomised controlled trial. Lancet 2015, 386, 444–451. [Google Scholar] [CrossRef]
  56. Ascierto, P.A.; McArthur, G.A.; Dréno, B.; Atkinson, V.; Liszkay, G.; Di Giacomo, A.M.; Mandalà, M.; Demidov, L.; Stroyakovskiy, D.; Thomas, L.; et al. Cobimetinib combined with vemurafenib in advanced BRAF(V600)-mutant melanoma (coBRIM): Updated efficacy results from a randomised, double-blind, phase 3 trial. Lancet Oncol. 2016, 17, 1248–1260. [Google Scholar] [CrossRef]
  57. Dummer, R.; Ascierto, P.A.; Gogas, H.J.; Arance, A.; Mandala, M.; Liszkay, G.; Garbe, C.; Schadendorf, D.; Krajsova, I.; Gutzmer, R.; et al. Encorafenib plus binimetinib versus vemurafenib or encorafenib in patients with BRAF-mutant melanoma (COLUMBUS): A multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2018, 19, 603–615. [Google Scholar] [CrossRef] [Green Version]
  58. Robert, C.; Karaszewska, B.; Schachter, J.; Rutkowski, P.; Mackiewicz, A.; Stroiakovski, D.; Lichinitser, M.; Dummer, R.; Grange, F.; Mortier, L.; et al. Improved overall survival in melanoma with combined dabrafenib and trametinib. N. Engl. J. Med. 2015, 372, 30–39. [Google Scholar] [CrossRef] [Green Version]
  59. Genomic Classification of Cutaneous Melanoma. Cell 2015, 161, 1681–1696. [CrossRef] [Green Version]
  60. Dummer, R.; Schadendorf, D.; Ascierto, P.A.; Arance, A.; Dutriaux, C.; Di Giacomo, A.M.; Rutkowski, P.; Del Vecchio, M.; Gutzmer, R.; Mandala, M.; et al. Binimetinib versus dacarbazine in patients with advanced NRAS-mutant melanoma (NEMO): A multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2017, 18, 435–445. [Google Scholar] [CrossRef]
  61. Lebbé, C.; Dutriaux, C.; Lesimple, T.; Kruit, W.; Kerger, J.; Thomas, L.; Guillot, B.; Braud, F.; Garbe, C.; Grob, J.J.; et al. Pimasertib Versus Dacarbazine in Patients With Unresectable NRAS-Mutated Cutaneous Melanoma: Phase II, Randomized, Controlled Trial with Crossover. Cancers 2020, 12, 1727. [Google Scholar] [CrossRef] [PubMed]
  62. Maertens, O.; Johnson, B.; Hollstein, P.; Frederick, D.T.; Cooper, Z.A.; Messiaen, L.; Bronson, R.T.; McMahon, M.; Granter, S.; Flaherty, K.; et al. Elucidating distinct roles for NF1 in melanomagenesis. Cancer Discov. 2013, 3, 338–349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Guo, J.; Si, L.; Kong, Y.; Flaherty, K.T.; Xu, X.; Zhu, Y.; Corless, C.L.; Li, L.; Li, H.; Sheng, X.; et al. Phase II, open-label, single-arm trial of imatinib mesylate in patients with metastatic melanoma harboring c-Kit mutation or amplification. J. Clin. Oncol. 2011, 29, 2904–2909. [Google Scholar] [CrossRef] [PubMed]
  64. Du, K.; Wei, S.; Wei, Z.; Frederick, D.T.; Miao, B.; Moll, T.; Tian, T.; Sugarman, E.; Gabrilovich, D.I.; Sullivan, R.J.; et al. Pathway signatures derived from on-treatment tumor specimens predict response to anti-PD1 blockade in metastatic melanoma. Nat. Commun. 2021, 12, 6023. [Google Scholar] [CrossRef]
  65. 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] [Green Version]
  66. Rizvi, N.A.; Hellmann, M.D.; Snyder, A.; Kvistborg, P.; Makarov, V.; Havel, J.J.; Lee, W.; Yuan, J.; Wong, P.; Ho, T.S.; et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015, 348, 124–128. [Google Scholar] [CrossRef] [Green Version]
  67. 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, eaar3593. [Google Scholar] [CrossRef] [Green Version]
  68. Topalian, S.L.; Taube, J.M.; Anders, R.A.; Pardoll, D.M. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat. Rev. Cancer 2016, 16, 275–287. [Google Scholar] [CrossRef]
  69. McGranahan, N.; Furness, A.J.; Rosenthal, R.; Ramskov, S.; Lyngaa, R.; Saini, S.K.; Jamal-Hanjani, M.; Wilson, G.A.; Birkbak, N.J.; Hiley, C.T.; et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016, 351, 1463–1469. [Google Scholar] [CrossRef] [Green Version]
  70. Chowell, D.; Morris, L.G.T.; Grigg, C.M.; Weber, J.K.; Samstein, R.M.; Makarov, V.; Kuo, F.; Kendall, S.M.; Requena, D.; Riaz, N.; et al. Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science 2018, 359, 582–587. [Google Scholar] [CrossRef] [Green Version]
  71. Paulson, K.G.; Voillet, V.; McAfee, M.S.; Hunter, D.S.; Wagener, F.D.; Perdicchio, M.; Valente, W.J.; Koelle, S.J.; Church, C.D.; Vandeven, N.; et al. Acquired cancer resistance to combination immunotherapy from transcriptional loss of class I HLA. Nat. Commun. 2018, 9, 3868. [Google Scholar] [CrossRef] [PubMed]
  72. Rooney, M.S.; Shukla, S.A.; Wu, C.J.; Getz, G.; Hacohen, N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell 2015, 160, 48–61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Davoli, T.; Uno, H.; Wooten, E.C.; Elledge, S.J. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science 2017, 355, eaaf8399. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Carter, J.A.; Gilbo, P.; Atwal, G.S. IMPRES does not reproducibly predict response to immune checkpoint blockade therapy in metastatic melanoma. Nat. Med. 2019, 25, 1833–1835. [Google Scholar] [CrossRef]
  75. Auslander, N.; Lee, J.S.; Ruppin, E. Reply to: ‘IMPRES does not reproducibly predict response to immune checkpoint blockade therapy in metastatic melanoma’. Nat. Med. 2019, 25, 1836–1838. [Google Scholar] [CrossRef]
  76. 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]
  77. Xiao, X.; Xu, C.; Yang, W.; Yu, R. Inconsistent prediction capability of ImmuneCells.Sig across different RNA-seq datasets. Nat. Commun. 2021, 12, 4167. [Google Scholar] [CrossRef]
  78. Xiong, D.; Wang, Y.; You, M. Reply to: "Inconsistent prediction capability of ImmuneCells.Sig across different RNA-seq datasets". Nat. Commun. 2021, 12, 4168. [Google Scholar] [CrossRef]
  79. Xiong, D.; Wang, Y.; You, M. A gene expression signature of TREM2(hi) macrophages and gammadelta T cells predicts immunotherapy response. Nat. Commun. 2020, 11, 5084. [Google Scholar] [CrossRef]
  80. 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]
  81. 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] [PubMed]
  82. 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]
  83. 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-gamma-related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Invest. 2017, 127, 2930–2940. [Google Scholar] [CrossRef] [PubMed]
  84. Ock, C.Y.; Hwang, J.E.; Keam, B.; Kim, S.B.; Shim, J.J.; Jang, H.J.; Park, S.; Sohn, B.H.; Cha, M.; Ajani, J.A.; et al. Genomic landscape associated with potential response to anti-CTLA-4 treatment in cancers. Nat. Commun. 2017, 8, 1050. [Google Scholar] [CrossRef] [Green Version]
  85. 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] [Green Version]
  86. Jerby-Arnon, L.; Shah, P.; Cuoco, M.S.; Rodman, C.; Su, M.J.; Melms, J.C.; Leeson, R.; Kanodia, A.; Mei, S.; Lin, J.R.; et al. A Cancer Cell Program Promotes T Cell Exclusion and Resistance to Checkpoint Blockade. Cell 2018, 175, 984–997.e24. [Google Scholar] [CrossRef] [Green Version]
  87. 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] [Green Version]
  88. 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, eaar3342. [Google Scholar] [CrossRef] [Green Version]
  89. Ben-Hamo, R.; Jacob Berger, A.; Gavert, N.; Miller, M.; Pines, G.; Oren, R.; Pikarsky, E.; Benes, C.H.; Neuman, T.; Zwang, Y.; et al. Predicting and affecting response to cancer therapy based on pathway-level biomarkers. Nat. Commun. 2020, 11, 3296. [Google Scholar] [CrossRef]
  90. Bild, A.H.; Yao, G.; Chang, J.T.; Wang, Q.; Potti, A.; Chasse, D.; Joshi, M.B.; Harpole, D.; Lancaster, J.M.; Berchuck, A.; et al. Oncogenic pathway signatures in human cancers as a guide to targeted therapies. Nature 2006, 439, 353–357. [Google Scholar] [CrossRef]
  91. Tian, S.; Simon, I.; Moreno, V.; Roepman, P.; Tabernero, J.; Snel, M.; van’t Veer, L.; Salazar, R.; Bernards, R.; Capella, G. A combined oncogenic pathway signature of BRAF, KRAS and PI3KCA mutation improves colorectal cancer classification and cetuximab treatment prediction. Gut 2013, 62, 540–549. [Google Scholar] [CrossRef] [PubMed]
  92. Haider, S.; Yao, C.Q.; Sabine, V.S.; Grzadkowski, M.; Stimper, V.; Starmans, M.H.W.; Wang, J.; Nguyen, F.; Moon, N.C.; Lin, X.; et al. Pathway-based subnetworks enable cross-disease biomarker discovery. Nat. Commun. 2018, 9, 4746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Grasso, C.S.; Tsoi, J.; Onyshchenko, M.; Abril-Rodriguez, G.; Ross-Macdonald, P.; Wind-Rotolo, M.; Champhekar, A.; Medina, E.; Torrejon, D.Y.; Shin, D.S.; et al. Conserved Interferon-gamma Signaling Drives Clinical Response to Immune Checkpoint Blockade Therapy in Melanoma. Cancer Cell 2020, 38, 500–515.e3. [Google Scholar] [CrossRef] [PubMed]
  94. Cabrita, R.; Lauss, M.; Sanna, A.; Donia, M.; Skaarup Larsen, M.; Mitra, S.; Johansson, I.; Phung, B.; Harbst, K.; Vallon-Christersson, J.; et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature 2020, 577, 561–565. [Google Scholar] [CrossRef]
  95. Fehrenbacher, L.; Spira, A.; Ballinger, M.; Kowanetz, M.; Vansteenkiste, J.; Mazieres, J.; Park, K.; Smith, D.; Artal-Cortes, A.; Lewanski, C.; et al. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): A multicentre, open-label, phase 2 randomised controlled trial. Lancet 2016, 387, 1837–1846. [Google Scholar] [CrossRef]
  96. 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] [Green Version]
  97. Petitprez, F.; de Reyniès, A.; Keung, E.Z.; Chen, T.W.; Sun, C.M.; Calderaro, J.; Jeng, Y.M.; Hsiao, L.P.; Lacroix, L.; Bougoüin, A.; et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature 2020, 577, 556–560. [Google Scholar] [CrossRef]
  98. Sade-Feldman, M.; Yizhak, K.; Bjorgaard, S.L.; Ray, J.P.; de Boer, C.G.; Jenkins, R.W.; Lieb, D.J.; Chen, J.H.; Frederick, D.T.; Barzily-Rokni, M.; et al. Defining T Cell States Associated with Response to Checkpoint Immunotherapy in Melanoma. Cell 2018, 175, 998–1013.e20. [Google Scholar] [CrossRef] [Green Version]
  99. Bownes, R.J.; Turnbull, A.K.; Martinez-Perez, C.; Cameron, D.A.; Sims, A.H.; Oikonomidou, O. On-treatment biomarkers can improve prediction of response to neoadjuvant chemotherapy in breast cancer. Breast Cancer Res. 2019, 21, 73. [Google Scholar] [CrossRef] [Green Version]
  100. Turnbull, A.K.; Arthur, L.M.; Renshaw, L.; Larionov, A.A.; Kay, C.; Dunbier, A.K.; Thomas, J.S.; Dowsett, M.; Sims, A.H.; Dixon, J.M. Accurate Prediction and Validation of Response to Endocrine Therapy in Breast Cancer. J. Clin. Oncol. 2015, 33, 2270–2278. [Google Scholar] [CrossRef]
  101. Ellis, M.J.; Suman, V.J.; Hoog, J.; Goncalves, R.; Sanati, S.; Creighton, C.J.; DeSchryver, K.; Crouch, E.; Brink, A.; Watson, M.; et al. Ki67 Proliferation Index as a Tool for Chemotherapy Decisions During and After Neoadjuvant Aromatase Inhibitor Treatment of Breast Cancer: Results From the American College of Surgeons Oncology Group Z1031 Trial (Alliance). J. Clin. Oncol. 2017, 35, 1061–1069. [Google Scholar] [CrossRef] [PubMed]
  102. Wallin, J.J.; Bendell, J.C.; Funke, R.; Sznol, M.; Korski, K.; Jones, S.; Hernandez, G.; Mier, J.; He, X.; Hodi, F.S.; et al. Atezolizumab in combination with bevacizumab enhances antigen-specific T-cell migration in metastatic renal cell carcinoma. Nat. Commun. 2016, 7, 12624. [Google Scholar] [CrossRef] [Green Version]
  103. Harel, M.; Ortenberg, R.; Varanasi, S.K.; Mangalhara, K.C.; Mardamshina, M.; Markovits, E.; Baruch, E.N.; Tripple, V.; Arama-Chayoth, M.; Greenberg, E.; et al. Proteomics of Melanoma Response to Immunotherapy Reveals Mitochondrial Dependence. Cell 2019, 179, 236–250.e18. [Google Scholar] [CrossRef]
  104. Lee, J.H.; Shklovskaya, E.; Lim, S.Y.; Carlino, M.S.; Menzies, A.M.; Stewart, A.; Pedersen, B.; Irvine, M.; Alavi, S.; Yang, J.Y.H.; et al. Transcriptional downregulation of MHC class I and melanoma de-differentiation in resistance to PD-1 inhibition. Nat. Commun. 2020, 11, 1897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Vogelstein, B.; Papadopoulos, N.; Velculescu, V.E.; Zhou, S.; Diaz, L.A., Jr.; Kinzler, K.W. Cancer genome landscapes. Science 2013, 339, 1546–1558. [Google Scholar] [CrossRef]
  106. Leko, V.; Rosenberg, S.A. Identifying and Targeting Human Tumor Antigens for T Cell-Based Immunotherapy of Solid Tumors. Cancer Cell 2020, 38, 454–472. [Google Scholar] [CrossRef] [PubMed]
  107. Turajlic, S.; Litchfield, K.; Xu, H.; Rosenthal, R.; McGranahan, N.; Reading, J.L.; Wong, Y.N.S.; Rowan, A.; Kanu, N.; Al Bakir, M.; et al. Insertion-and-deletion-derived tumour-specific neoantigens and the immunogenic phenotype: A pan-cancer analysis. Lancet Oncol. 2017, 18, 1009–1021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Coulie, P.G.; Van den Eynde, B.J.; van der Bruggen, P.; Boon, T. Tumour antigens recognized by T lymphocytes: At the core of cancer immunotherapy. Nat. Rev. Cancer 2014, 14, 135–146. [Google Scholar] [CrossRef] [PubMed]
  109. Roszik, J.; Wang, W.L.; Livingston, J.A.; Roland, C.L.; Ravi, V.; Yee, C.; Hwu, P.; Futreal, A.; Lazar, A.J.; Patel, S.R.; et al. Overexpressed PRAME is a potential immunotherapy target in sarcoma subtypes. Clin. Sarcoma Res. 2017, 7, 11. [Google Scholar] [CrossRef] [Green Version]
  110. Zhang, W.; Li, L.; Cai, L.; Liang, Y.; Xu, J.; Liu, Y.; Zhou, L.; Ding, C.; Zhang, Y.; Zhao, H.; et al. Tumor-associated antigen Prame targets tumor suppressor p14/ARF for degradation as the receptor protein of CRL2(Prame) complex. Cell Death Differ. 2021, 28, 1926–1940. [Google Scholar] [CrossRef]
  111. Smyth, M.J.; Ngiow, S.F.; Ribas, A.; Teng, M.W. Combination cancer immunotherapies tailored to the tumour microenvironment. Nat. Rev. Clin. Oncol. 2016, 13, 143–158. [Google Scholar] [CrossRef] [PubMed]
  112. Hogan, S.A.; Levesque, M.P.; Cheng, P.F. Melanoma Immunotherapy: Next-Generation Biomarkers. Front. Oncol. 2018, 8, 178. [Google Scholar] [CrossRef]
  113. Keenan, T.E.; Burke, K.P.; Van Allen, E.M. Genomic correlates of response to immune checkpoint blockade. Nat. Med. 2019, 25, 389–402. [Google Scholar] [CrossRef] [PubMed]
  114. Trujillo, J.A.; Luke, J.J.; Zha, Y.; Segal, J.P.; Ritterhouse, L.L.; Spranger, S.; Matijevich, K.; Gajewski, T.F. Secondary resistance to immunotherapy associated with beta-catenin pathway activation or PTEN loss in metastatic melanoma. J. Immunother. Cancer 2019, 7, 295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Curran, M.A.; Montalvo, W.; Yagita, H.; Allison, J.P. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc. Natl. Acad. Sci. USA 2010, 107, 4275–4280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Sharma, P.; Allison, J.P. The future of immune checkpoint therapy. Science 2015, 348, 56–61. [Google Scholar] [CrossRef] [PubMed]
  117. Wei, S.C.; Levine, J.H.; Cogdill, A.P.; Zhao, Y.; Anang, N.A.S.; Andrews, M.C.; Sharma, P.; Wang, J.; Wargo, J.A.; Pe’er, D.; et al. Distinct Cellular Mechanisms Underlie Anti-CTLA-4 and Anti-PD-1 Checkpoint Blockade. Cell 2017, 170, 1120–1133.e17. [Google Scholar] [CrossRef] [Green Version]
  118. Ribas, A.; Wolchok, J.D. Cancer immunotherapy using checkpoint blockade. Science 2018, 359, 1350–1355. [Google Scholar] [CrossRef] [Green Version]
  119. Herbst, R.S.; Soria, J.C.; Kowanetz, M.; Fine, G.D.; Hamid, O.; Gordon, M.S.; Sosman, J.A.; McDermott, D.F.; Powderly, J.D.; Gettinger, S.N.; et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014, 515, 563–567. [Google Scholar] [CrossRef] [Green Version]
  120. Taube, J.M.; Klein, A.; Brahmer, J.R.; Xu, H.; Pan, X.; Kim, J.H.; Chen, L.; Pardoll, D.M.; Topalian, S.L.; Anders, R.A. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin. Cancer Res. 2014, 20, 5064–5074. [Google Scholar] [CrossRef] [Green Version]
  121. Tumeh, P.C.; Harview, C.L.; Yearley, J.H.; Shintaku, I.P.; Taylor, E.J.; 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]
  122. Sharma, A.; Subudhi, S.K.; Blando, J.; Vence, L.; Wargo, J.; Allison, J.P.; Ribas, A.; Sharma, P. Anti-CTLA-4 Immunotherapy Does Not Deplete FOXP3(+) Regulatory T Cells (Tregs) in Human Cancers-Response. Clin. Cancer Res. 2019, 25, 3469–3470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Shen, H.; Huang, F.; Zhang, X.; Ojo, O.A.; Li, Y.; Trummell, H.Q.; Anderson, J.C.; Fiveash, J.; Bredel, M.; Yang, E.S.; et al. Selective suppression of melanoma lacking IFN-gamma pathway by JAK inhibition depends on T cells and host TNF signaling. Nat. Commun. 2022, 13, 5013. [Google Scholar] [CrossRef] [PubMed]
  124. Liu, Y.T.; Sun, Z.J. Turning cold tumors into hot tumors by improving T-cell infiltration. Theranostics 2021, 11, 5365–5386. [Google Scholar] [CrossRef] [PubMed]
  125. Chen, D.S.; Mellman, I. Oncology meets immunology: The cancer-immunity cycle. Immunity 2013, 39, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Chen, D.S.; Mellman, I. Elements of cancer immunity and the cancer-immune set point. Nature 2017, 541, 321–330. [Google Scholar] [CrossRef] [PubMed]
  127. Hegde, P.S.; Karanikas, V.; Evers, S. The Where, the When, and the How of Immune Monitoring for Cancer Immunotherapies in the Era of Checkpoint Inhibition. Clin. Cancer Res. 2016, 22, 1865–1874. [Google Scholar] [CrossRef] [Green Version]
  128. Galon, J.; Bruni, D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat. Rev. Drug Discov. 2019, 18, 197–218. [Google Scholar] [CrossRef]
  129. Hegde, P.S.; Chen, D.S. Top 10 Challenges in Cancer Immunotherapy. Immunity 2020, 52, 17–35. [Google Scholar] [CrossRef]
  130. Ansell, S.M.; Lesokhin, A.M.; Borrello, I.; Halwani, A.; Scott, E.C.; Gutierrez, M.; Schuster, S.J.; Millenson, M.M.; Cattry, D.; Freeman, G.J.; et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N. Engl. J. Med. 2015, 372, 311–319. [Google Scholar] [CrossRef] [Green Version]
  131. Gubin, M.M.; Zhang, X.; Schuster, H.; Caron, E.; Ward, J.P.; Noguchi, T.; Ivanova, Y.; Hundal, J.; Arthur, C.D.; Krebber, W.J.; et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 2014, 515, 577–581. [Google Scholar] [CrossRef] [PubMed]
  132. Marincola, F.M.; Jaffee, E.M.; Hicklin, D.J.; Ferrone, S. Escape of human solid tumors from T-cell recognition: Molecular mechanisms and functional significance. Adv. Immunol. 2000, 74, 181–273. [Google Scholar] [PubMed]
  133. Sucker, A.; Zhao, F.; Real, B.; Heeke, C.; Bielefeld, N.; Maβen, S.; Horn, S.; Moll, I.; Maltaner, R.; Horn, P.A.; et al. Genetic evolution of T-cell resistance in the course of melanoma progression. Clin. Cancer Res. 2014, 20, 6593–6604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Liu, C.; Peng, W.; Xu, C.; Lou, Y.; Zhang, M.; Wargo, J.A.; Chen, J.Q.; Li, H.S.; Watowich, S.S.; Yang, Y.; et al. BRAF inhibition increases tumor infiltration by T cells and enhances the antitumor activity of adoptive immunotherapy in mice. Clin. Cancer Res. 2013, 19, 393–403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Spranger, S.; Bao, R.; Gajewski, T.F. Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity. Nature 2015, 523, 231–235. [Google Scholar] [CrossRef]
  136. Shin, D.S.; Zaretsky, J.M.; Escuin-Ordinas, H.; Garcia-Diaz, A.; Hu-Lieskovan, S.; Kalbasi, A.; Grasso, C.S.; Hugo, W.; Sandoval, S.; Torrejon, D.Y.; et al. Primary Resistance to PD-1 Blockade Mediated by JAK1/2 Mutations. Cancer Discov. 2017, 7, 188–201. [Google Scholar] [CrossRef] [Green Version]
  137. He, Y.; Cao, J.; Zhao, C.; Li, X.; Zhou, C.; Hirsch, F.R. TIM-3, a promising target for cancer immunotherapy. Onco. Targets Ther. 2018, 11, 7005–7009. [Google Scholar] [CrossRef] [Green Version]
  138. Kryczek, I.; Zou, L.; Rodriguez, P.; Zhu, G.; Wei, S.; Mottram, P.; Brumlik, M.; Cheng, P.; Curiel, T.; Myers, L.; et al. B7-H4 expression identifies a novel suppressive macrophage population in human ovarian carcinoma. J. Exp. Med. 2006, 203, 871–881. [Google Scholar] [CrossRef]
  139. Kuang, D.M.; Zhao, Q.; Peng, C.; Xu, J.; Zhang, J.P.; Wu, C.; Zheng, L. Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J. Exp. Med. 2009, 206, 1327–1337. [Google Scholar] [CrossRef] [Green Version]
  140. 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]
  141. Şenbabaoğlu, Y.; Gejman, R.S.; Winer, A.G.; Liu, M.; Van Allen, E.M.; de Velasco, G.; Miao, D.; Ostrovnaya, I.; Drill, E.; Luna, A.; et al. Tumor immune microenvironment characterization in clear cell renal cell carcinoma identifies prognostic and immunotherapeutically relevant messenger RNA signatures. Genome Biol. 2016, 17, 231. [Google Scholar] [CrossRef] [PubMed]
  142. Charoentong, P.; Finotello, F.; Angelova, M.; Mayer, C.; Efremova, M.; Rieder, D.; Hackl, H.; Trajanoski, Z. Pan-cancer Immunogenomic Analyses Reveal Genotype-Immunophenotype Relationships and Predictors of Response to Checkpoint Blockade. Cell Rep. 2017, 18, 248–262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Prat, A.; Navarro, A.; Paré, L.; Reguart, N.; Galván, P.; Pascual, T.; Martínez, A.; Nuciforo, P.; Comerma, L.; Alos, L.; et al. Immune-Related Gene Expression Profiling After PD-1 Blockade in Non-Small Cell Lung Carcinoma, Head and Neck Squamous Cell Carcinoma, and Melanoma. Cancer Res. 2017, 77, 3540–3550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Danaher, P.; Warren, S.; Lu, R.; Samayoa, J.; Sullivan, A.; Pekker, I.; Wallden, B.; Marincola, F.M.; Cesano, A. Pan-cancer adaptive immune resistance as defined by the Tumor Inflammation Signature (TIS): Results from The Cancer Genome Atlas (TCGA). J. Immunother. Cancer 2018, 6, 63. [Google Scholar] [CrossRef] [Green Version]
  145. McDermott, D.F.; Huseni, M.A.; Atkins, M.B.; Motzer, R.J.; Rini, B.I.; Escudier, B.; Fong, L.; Joseph, R.W.; Pal, S.K.; Reeves, J.A.; et al. Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma. Nat. Med. 2018, 24, 749–757. [Google Scholar] [CrossRef]
  146. Anagnostou, V.; Smith, K.N.; Forde, P.M.; Niknafs, N.; Bhattacharya, R.; White, J.; Zhang, T.; Adleff, V.; Phallen, J.; Wali, N.; et al. Evolution of Neoantigen Landscape during Immune Checkpoint Blockade in Non-Small Cell Lung Cancer. Cancer Discov. 2017, 7, 264–276. [Google Scholar] [CrossRef] [Green Version]
  147. Im, S.J.; Hashimoto, M.; Gerner, M.Y.; Lee, J.; Kissick, H.T.; Burger, M.C.; Shan, Q.; Hale, J.S.; Lee, J.; Nasti, T.H.; et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 2016, 537, 417–421. [Google Scholar] [CrossRef] [Green Version]
  148. Seiwert, T.Y.; Burtness, B.; Mehra, R.; Weiss, J.; Berger, R.; Eder, J.P.; Heath, K.; McClanahan, T.; Lunceford, J.; Gause, C.; et al. Safety and clinical activity of pembrolizumab for treatment of recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-012): An open-label, multicentre, phase 1b trial. Lancet Oncol. 2016, 17, 956–965. [Google Scholar] [CrossRef]
  149. Fehlings, M.; Simoni, Y.; Penny, H.L.; Becht, E.; Loh, C.Y.; Gubin, M.M.; Ward, J.P.; Wong, S.C.; Schreiber, R.D.; Newell, E.W. Checkpoint blockade immunotherapy reshapes the high-dimensional phenotypic heterogeneity of murine intratumoural neoantigen-specific CD8(+) T cells. Nat. Commun. 2017, 8, 562. [Google Scholar] [CrossRef]
  150. van Rooij, N.; van Buuren, M.M.; Philips, D.; Velds, A.; Toebes, M.; Heemskerk, B.; van Dijk, L.J.; Behjati, S.; Hilkmann, H.; El Atmioui, D.; et al. Tumor exome analysis reveals neoantigen-specific T-cell reactivity in an ipilimumab-responsive melanoma. J. Clin. Oncol. 2013, 31, e439–e442. [Google Scholar] [CrossRef]
  151. Le, D.T.; Durham, J.N.; Smith, K.N.; Wang, H.; Bartlett, B.R.; Aulakh, L.K.; Lu, S.; Kemberling, H.; Wilt, C.; Luber, B.S.; et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017, 357, 409–413. [Google Scholar] [CrossRef] [PubMed]
  152. Le, D.T.; Uram, J.N.; Wang, H.; Bartlett, B.R.; Kemberling, H.; Eyring, A.D.; Skora, A.D.; Luber, B.S.; Azad, N.S.; Laheru, D.; et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N. Engl. J. Med. 2015, 372, 2509–2520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Nghiem, P.T.; Bhatia, S.; Lipson, E.J.; Kudchadkar, R.R.; Miller, N.J.; Annamalai, L.; Berry, S.; Chartash, E.K.; Daud, A.; Fling, S.P.; et al. PD-1 Blockade with Pembrolizumab in Advanced Merkel-Cell Carcinoma. N. Engl. J. Med. 2016, 374, 2542–2552. [Google Scholar] [CrossRef] [PubMed]
  154. Łuksza, M.; Riaz, N.; Makarov, V.; Balachandran, V.P.; Hellmann, M.D.; Solovyov, A.; Rizvi, N.A.; Merghoub, T.; Levine, A.J.; Chan, T.A.; et al. A neoantigen fitness model predicts tumour response to checkpoint blockade immunotherapy. Nature 2017, 551, 517–520. [Google Scholar] [CrossRef]
  155. Panda, A.; de Cubas, A.A.; Stein, M.; Riedlinger, G.; Kra, J.; Mayer, T.; Smith, C.C.; Vincent, B.G.; Serody, J.S.; Beckermann, K.E.; et al. Endogenous retrovirus expression is associated with response to immune checkpoint blockade in clear cell renal cell carcinoma. JCI Insight 2018, 3, e121522. [Google Scholar] [CrossRef]
  156. Rosenberg, J.E.; Hoffman-Censits, J.; Powles, T.; van der Heijden, M.S.; Balar, A.V.; Necchi, A.; Dawson, N.; O’Donnell, P.H.; Balmanoukian, A.; Loriot, Y.; et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: A single-arm, multicentre, phase 2 trial. Lancet 2016, 387, 1909–1920. [Google Scholar] [CrossRef] [Green Version]
  157. Hellmann, M.D.; Callahan, M.K.; Awad, M.M.; Calvo, E.; Ascierto, P.A.; Atmaca, A.; Rizvi, N.A.; Hirsch, F.R.; Selvaggi, G.; Szustakowski, J.D.; et al. Tumor Mutational Burden and Efficacy of Nivolumab Monotherapy and in Combination with Ipilimumab in Small-Cell Lung Cancer. Cancer Cell 2018, 33, 853–861.e4. [Google Scholar] [CrossRef] [Green Version]
  158. 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]
  159. Miao, D.; Margolis, C.A.; Vokes, N.I.; Liu, D.; Taylor-Weiner, A.; Wankowicz, S.M.; Adeegbe, D.; Keliher, D.; Schilling, B.; Tracy, A.; et al. Genomic correlates of response to immune checkpoint blockade in microsatellite-stable solid tumors. Nat. Genet. 2018, 50, 1271–1281. [Google Scholar] [CrossRef]
  160. Wang, S.; Jia, M.; He, Z.; Liu, X.S. APOBEC3B and APOBEC mutational signature as potential predictive markers for immunotherapy response in non-small cell lung cancer. Oncogene 2018, 37, 3924–3936. [Google Scholar] [CrossRef]
  161. Chen, R.; Zinzani, P.L.; Fanale, M.A.; Armand, P.; Johnson, N.A.; Brice, P.; Radford, J.; Ribrag, V.; Molin, D.; Vassilakopoulos, T.P.; et al. Phase II Study of the Efficacy and Safety of Pembrolizumab for Relapsed/Refractory Classic Hodgkin Lymphoma. J. Clin. Oncol. 2017, 35, 2125–2132. [Google Scholar] [CrossRef] [PubMed]
  162. Green, M.R.; Monti, S.; Rodig, S.J.; Juszczynski, P.; Currie, T.; O’Donnell, E.; Chapuy, B.; Takeyama, K.; Neuberg, D.; Golub, T.R.; et al. Integrative analysis reveals selective 9p24.1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma. Blood 2010, 116, 3268–3277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Nayak, L.; Iwamoto, F.M.; LaCasce, A.; Mukundan, S.; Roemer, M.G.M.; Chapuy, B.; Armand, P.; Rodig, S.J.; Shipp, M.A. PD-1 blockade with nivolumab in relapsed/refractory primary central nervous system and testicular lymphoma. Blood 2017, 129, 3071–3073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Goodman, A.M.; Piccioni, D.; Kato, S.; Boichard, A.; Wang, H.Y.; Frampton, G.; Lippman, S.M.; Connelly, C.; Fabrizio, D.; Miller, V.; et al. Prevalence of PDL1 Amplification and Preliminary Response to Immune Checkpoint Blockade in Solid Tumors. JAMA Oncol. 2018, 4, 1237–1244. [Google Scholar] [CrossRef] [Green Version]
  165. Zhang, J.; Bu, X.; Wang, H.; Zhu, Y.; Geng, Y.; Nihira, N.T.; Tan, Y.; Ci, Y.; Wu, F.; Dai, X.; et al. Cyclin D-CDK4 kinase destabilizes PD-L1 via cullin 3-SPOP to control cancer immune surveillance. Nature 2018, 553, 91–95. [Google Scholar] [CrossRef] [Green Version]
  166. Mezzadra, R.; Sun, C.; Jae, L.T.; Gomez-Eerland, R.; de Vries, E.; Wu, W.; Logtenberg, M.E.W.; Slagter, M.; Rozeman, E.A.; Hofland, I.; et al. Identification of CMTM6 and CMTM4 as PD-L1 protein regulators. Nature 2017, 549, 106–110. [Google Scholar] [CrossRef]
  167. Burr, M.L.; Sparbier, C.E.; Chan, Y.C.; Williamson, J.C.; Woods, K.; Beavis, P.A.; Lam, E.Y.N.; Henderson, M.A.; Bell, C.C.; Stolzenburg, S.; et al. CMTM6 maintains the expression of PD-L1 and regulates anti-tumour immunity. Nature 2017, 549, 101–105. [Google Scholar] [CrossRef] [Green Version]
  168. Pan, D.; Kobayashi, A.; Jiang, P.; Ferrari de Andrade, L.; Tay, R.E.; Luoma, A.M.; Tsoucas, D.; Qiu, X.; Lim, K.; Rao, P.; et al. A major chromatin regulator determines resistance of tumor cells to T cell-mediated killing. Science 2018, 359, 770–775. [Google Scholar] [CrossRef] [Green Version]
  169. Shen, J.; Ju, Z.; Zhao, W.; Wang, L.; Peng, Y.; Ge, Z.; Nagel, Z.D.; Zou, J.; Wang, C.; Kapoor, P.; et al. ARID1A deficiency promotes mutability and potentiates therapeutic antitumor immunity unleashed by immune checkpoint blockade. Nat. Med. 2018, 24, 556–562. [Google Scholar] [CrossRef]
  170. Jelinic, P.; Ricca, J.; Van Oudenhove, E.; Olvera, N.; Merghoub, T.; Levine, D.A.; Zamarin, D. Immune-Active Microenvironment in Small Cell Carcinoma of the Ovary, Hypercalcemic Type: Rationale for Immune Checkpoint Blockade. J. Natl. Cancer Inst. 2018, 110, 787–790. [Google Scholar] [CrossRef] [Green Version]
  171. Shukla, S.A.; Bachireddy, P.; Schilling, B.; Galonska, C.; Zhan, Q.; Bango, C.; Langer, R.; Lee, P.C.; Gusenleitner, D.; Keskin, D.B.; et al. Cancer-Germline Antigen Expression Discriminates Clinical Outcome to CTLA-4 Blockade. Cell 2018, 173, 624–633.e8. [Google Scholar] [CrossRef] [PubMed]
  172. Rizvi, H.; Sanchez-Vega, F.; La, K.; Chatila, W.; Jonsson, P.; Halpenny, D.; Plodkowski, A.; Long, N.; Sauter, J.L.; Rekhtman, N.; et al. Molecular Determinants of Response to Anti-Programmed Cell Death (PD)-1 and Anti-Programmed Death-Ligand 1 (PD-L1) Blockade in Patients With Non-Small-Cell Lung Cancer Profiled With Targeted Next-Generation Sequencing. J. Clin. Oncol. 2018, 36, 633–641. [Google Scholar] [CrossRef] [PubMed]
  173. Koyama, S.; Akbay, E.A.; Li, Y.Y.; Aref, A.R.; Skoulidis, F.; Herter-Sprie, G.S.; Buczkowski, K.A.; Liu, Y.; Awad, M.M.; Denning, W.L.; et al. STK11/LKB1 Deficiency Promotes Neutrophil Recruitment and Proinflammatory Cytokine Production to Suppress T-cell Activity in the Lung Tumor Microenvironment. Cancer Res. 2016, 76, 999–1008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Peng, W.; Chen, J.Q.; Liu, C.; Malu, S.; Creasy, C.; Tetzlaff, M.T.; Xu, C.; McKenzie, J.A.; Zhang, C.; Liang, X.; et al. Loss of PTEN Promotes Resistance to T Cell-Mediated Immunotherapy. Cancer Discov. 2016, 6, 202–216. [Google Scholar] [CrossRef] [Green Version]
  175. George, S.; Miao, D.; Demetri, G.D.; Adeegbe, D.; Rodig, S.J.; Shukla, S.; Lipschitz, M.; Amin-Mansour, A.; Raut, C.P.; Carter, S.L.; et al. Loss of PTEN Is Associated with Resistance to Anti-PD-1 Checkpoint Blockade Therapy in Metastatic Uterine Leiomyosarcoma. Immunity 2017, 46, 197–204. [Google Scholar] [CrossRef] [Green Version]
  176. Spranger, S.; Dai, D.; Horton, B.; Gajewski, T.F. Tumor-Residing Batf3 Dendritic Cells Are Required for Effector T Cell Trafficking and Adoptive T Cell Therapy. Cancer Cell 2017, 31, 711–723.e4. [Google Scholar] [CrossRef] [Green Version]
  177. Gainor, J.F.; Shaw, A.T.; Sequist, L.V.; Fu, X.; Azzoli, C.G.; Piotrowska, Z.; Huynh, T.G.; Zhao, L.; Fulton, L.; Schultz, K.R.; et al. EGFR Mutations and ALK Rearrangements Are Associated with Low Response Rates to PD-1 Pathway Blockade in Non-Small Cell Lung Cancer: A Retrospective Analysis. Clin. Cancer Res. 2016, 22, 4585–4593. [Google Scholar] [CrossRef] [Green Version]
  178. Lee, C.K.; Man, J.; Lord, S.; Cooper, W.; Links, M.; Gebski, V.; Herbst, R.S.; Gralla, R.J.; Mok, T.; Yang, J.C. Clinical and Molecular Characteristics Associated With Survival Among Patients Treated With Checkpoint Inhibitors for Advanced Non-Small Cell Lung Carcinoma: A Systematic Review and Meta-analysis. JAMA Oncol. 2018, 4, 210–216. [Google Scholar] [CrossRef]
  179. Dong, Z.Y.; Zhong, W.Z.; Zhang, X.C.; Su, J.; Xie, Z.; Liu, S.Y.; Tu, H.Y.; Chen, H.J.; Sun, Y.L.; Zhou, Q.; et al. Potential Predictive Value of TP53 and KRAS Mutation Status for Response to PD-1 Blockade Immunotherapy in Lung Adenocarcinoma. Clin. Cancer Res. 2017, 23, 3012–3024. [Google Scholar] [CrossRef] [Green Version]
  180. Mariathasan, S.; Turley, S.J.; Nickles, D.; Castiglioni, A.; Yuen, K.; Wang, Y.; Kadel, E.E., III; Koeppen, H.; Astarita, J.L.; Cubas, R.; et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018, 554, 544–548. [Google Scholar] [CrossRef]
  181. Chakravarthy, A.; Khan, L.; Bensler, N.P.; Bose, P.; De Carvalho, D.D. TGF-β-associated extracellular matrix genes link cancer-associated fibroblasts to immune evasion and immunotherapy failure. Nat. Commun. 2018, 9, 4692. [Google Scholar] [CrossRef] [PubMed]
  182. O’Donnell, J.S.; Long, G.V.; Scolyer, R.A.; Teng, M.W.; Smyth, M.J. Resistance to PD1/PDL1 checkpoint inhibition. Cancer Treat. Rev. 2017, 52, 71–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Jenkins, R.W.; Barbie, D.A.; Flaherty, K.T. Mechanisms of resistance to immune checkpoint inhibitors. Br. J. Cancer 2018, 118, 9–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Cui, Y.; Guo, G. Immunomodulatory Function of the Tumor Suppressor p53 in Host Immune Response and the Tumor Microenvironment. Int. J. Mol. Sci. 2016, 17, 1942. [Google Scholar] [CrossRef] [Green Version]
  185. Zhang, J.; Veeramachaneni, N. Targeting interleukin-1beta and inflammation in lung cancer. Biomark Res. 2022, 10, 5. [Google Scholar] [CrossRef]
  186. Zippelius, A.; Batard, P.; Rubio-Godoy, V.; Bioley, G.; Lienard, D.; Lejeune, F.; Rimoldi, D.; Guillaume, P.; Meidenbauer, N.; Mackensen, A.; et al. Effector function of human tumor-specific CD8 T cells in melanoma lesions: A state of local functional tolerance. Cancer Res. 2004, 64, 2865–2873. [Google Scholar] [CrossRef] [Green Version]
  187. Ribas, A.; Medina, T.; Kummar, S.; Amin, A.; Kalbasi, A.; Drabick, J.J.; Barve, M.; Daniels, G.A.; Wong, D.J.; Schmidt, E.V.; et al. SD-101 in Combination with Pembrolizumab in Advanced Melanoma: Results of a Phase Ib, Multicenter Study. Cancer Discov. 2018, 8, 1250–1257. [Google Scholar] [CrossRef] [Green Version]
  188. Chin, E.N.; Yu, C.; Vartabedian, V.F.; Jia, Y.; Kumar, M.; Gamo, A.M.; Vernier, W.; Ali, S.H.; Kissai, M.; Lazar, D.C.; et al. Antitumor activity of a systemic STING-activating non-nucleotide cGAMP mimetic. Science 2020, 369, 993–999. [Google Scholar] [CrossRef]
  189. Twumasi-Boateng, K.; Pettigrew, J.L.; Kwok, Y.Y.E.; Bell, J.C.; Nelson, B.H. Oncolytic viruses as engineering platforms for combination immunotherapy. Nat. Rev. Cancer 2018, 18, 419–432. [Google Scholar] [CrossRef]
  190. Ribas, A.; Dummer, R.; Puzanov, I.; VanderWalde, A.; Andtbacka, R.H.I.; Michielin, O.; Olszanski, A.J.; Malvehy, J.; Cebon, J.; Fernandez, E.; et al. Oncolytic Virotherapy Promotes Intratumoral T Cell Infiltration and Improves Anti-PD-1 Immunotherapy. Cell 2018, 174, 1031–1032. [Google Scholar] [CrossRef]
  191. Barker, H.E.; Paget, J.T.; Khan, A.A.; Harrington, K.J. The tumour microenvironment after radiotherapy: Mechanisms of resistance and recurrence. Nat. Rev. Cancer 2015, 15, 409–425. [Google Scholar] [CrossRef] [PubMed]
  192. Yamazaki, T.; Buqué, A.; Ames, T.D.; Galluzzi, L. PT-112 induces immunogenic cell death and synergizes with immune checkpoint blockers in mouse tumor models. Oncoimmunology 2020, 9, 1721810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Maatouk, D.M.; Kellam, L.D.; Mann, M.R.; Lei, H.; Li, E.; Bartolomei, M.S.; Resnick, J.L. DNA methylation is a primary mechanism for silencing postmigratory primordial germ cell genes in both germ cell and somatic cell lineages. Development 2006, 133, 3411–3418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Ritter, C.; Fan, K.; Paschen, A.; Reker Hardrup, S.; Ferrone, S.; Nghiem, P.; Ugurel, S.; Schrama, D.; Becker, J.C. Epigenetic priming restores the HLA class-I antigen processing machinery expression in Merkel cell carcinoma. Sci. Rep. 2017, 7, 2290. [Google Scholar] [CrossRef] [Green Version]
  195. Luo, N.; Nixon, M.J.; Gonzalez-Ericsson, P.I.; Sanchez, V.; Opalenik, S.R.; Li, H.; Zahnow, C.A.; Nickels, M.L.; Liu, F.; Tantawy, M.N.; et al. DNA methyltransferase inhibition upregulates MHC-I to potentiate cytotoxic T lymphocyte responses in breast cancer. Nat. Commun. 2018, 9, 248. [Google Scholar] [CrossRef] [Green Version]
  196. Seth, P.; Csizmadia, E.; Hedblom, A.; Vuerich, M.; Xie, H.; Li, M.; Longhi, M.S.; Wegiel, B. Deletion of Lactate Dehydrogenase-A in Myeloid Cells Triggers Antitumor Immunity. Cancer Res. 2017, 77, 3632–3643. [Google Scholar] [CrossRef] [Green Version]
  197. Pilon-Thomas, S.; Kodumudi, K.N.; El-Kenawi, A.E.; Russell, S.; Weber, A.M.; Luddy, K.; Damaghi, M.; Wojtkowiak, J.W.; Mulé, J.J.; Ibrahim-Hashim, A.; et al. Neutralization of Tumor Acidity Improves Antitumor Responses to Immunotherapy. Cancer Res. 2016, 76, 1381–1390. [Google Scholar] [CrossRef] [Green Version]
  198. Qi, X.; Yang, M.; Ma, L.; Sauer, M.; Avella, D.; Kaifi, J.T.; Bryan, J.; Cheng, K.; Staveley-O’Carroll, K.F.; Kimchi, E.T.; et al. Synergizing sunitinib and radiofrequency ablation to treat hepatocellular cancer by triggering the antitumor immune response. J. Immunother. Cancer 2020, 8, e001038. [Google Scholar] [CrossRef]
  199. Chen, Q.; He, Y.; Wang, Y.; Li, C.; Zhang, Y.; Guo, Q.; Zhang, Y.; Chu, Y.; Liu, P.; Chen, H.; et al. Penetrable Nanoplatform for "Cold" Tumor Immune Microenvironment Reeducation. Adv. Sci. (Weinh) 2020, 7, 2000411. [Google Scholar] [CrossRef]
  200. Sun, Y.; Zhang, Y.; Gao, Y.; Wang, P.; He, G.; Blum, N.T.; Lin, J.; Liu, Q.; Wang, X.; Huang, P. Six Birds with One Stone: Versatile Nanoporphyrin for Single-Laser-Triggered Synergistic Phototheranostics and Robust Immune Activation. Adv. Mater. 2020, 32, e2004481. [Google Scholar] [CrossRef]
  201. Zanganeh, S.; Hutter, G.; Spitler, R.; Lenkov, O.; Mahmoudi, M.; Shaw, A.; Pajarinen, J.S.; Nejadnik, H.; Goodman, S.; Moseley, M.; et al. Iron oxide nanoparticles inhibit tumour growth by inducing pro-inflammatory macrophage polarization in tumour tissues. Nat. Nanotechnol. 2016, 11, 986–994. [Google Scholar] [CrossRef] [PubMed]
  202. Liu, X.; Zheng, J.; Sun, W.; Zhao, X.; Li, Y.; Gong, N.; Wang, Y.; Ma, X.; Zhang, T.; Zhao, L.Y.; et al. Ferrimagnetic Vortex Nanoring-Mediated Mild Magnetic Hyperthermia Imparts Potent Immunological Effect for Treating Cancer Metastasis. ACS Nano 2019, 13, 8811–8825. [Google Scholar] [CrossRef] [PubMed]
  203. Chavez, M.; Silvestrini, M.T.; Ingham, E.S.; Fite, B.Z.; Mahakian, L.M.; Tam, S.M.; Ilovitsh, A.; Monjazeb, A.M.; Murphy, W.J.; Hubbard, N.E.; et al. Distinct immune signatures in directly treated and distant tumors result from TLR adjuvants and focal ablation. Theranostics 2018, 8, 3611–3628. [Google Scholar] [CrossRef] [PubMed]
  204. Ilovitsh, T.; Feng, Y.; Foiret, J.; Kheirolomoom, A.; Zhang, H.; Ingham, E.S.; Ilovitsh, A.; Tumbale, S.K.; Fite, B.Z.; Wu, B.; et al. Low-frequency ultrasound-mediated cytokine transfection enhances T cell recruitment at local and distant tumor sites. Proc. Natl. Acad. Sci. USA 2020, 117, 12674–12685. [Google Scholar] [CrossRef]
  205. Lai, J.; Mardiana, S.; House, I.G.; Sek, K.; Henderson, M.A.; Giuffrida, L.; Chen, A.X.Y.; Todd, K.L.; Petley, E.V.; Chan, J.D.; et al. Adoptive cellular therapy with T cells expressing the dendritic cell growth factor Flt3L drives epitope spreading and antitumor immunity. Nat. Immunol. 2020, 21, 914–926. [Google Scholar] [CrossRef]
  206. Rosenberg, S.A.; Yang, J.C.; Sherry, R.M.; Kammula, U.S.; Hughes, M.S.; Phan, G.Q.; Citrin, D.E.; Restifo, N.P.; Robbins, P.F.; Wunderlich, J.R.; et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin. Cancer Res. 2011, 17, 4550–4557. [Google Scholar] [CrossRef] [Green Version]
  207. Adachi, K.; Kano, Y.; Nagai, T.; Okuyama, N.; Sakoda, Y.; Tamada, K. IL-7 and CCL19 expression in CAR-T cells improves immune cell infiltration and CAR-T cell survival in the tumor. Nat. Biotechnol. 2018, 36, 346–351. [Google Scholar] [CrossRef]
  208. Ott, P.A.; Hu-Lieskovan, S.; Chmielowski, B.; Govindan, R.; Naing, A.; Bhardwaj, N.; Margolin, K.; Awad, M.M.; Hellmann, M.D.; Lin, J.J.; et al. A Phase Ib Trial of Personalized Neoantigen Therapy Plus Anti-PD-1 in Patients with Advanced Melanoma, Non-small Cell Lung Cancer, or Bladder Cancer. Cell 2020, 183, 347–362.e24. [Google Scholar] [CrossRef]
  209. Abril-Rodriguez, G.; Torrejon, D.Y.; Liu, W.; Zaretsky, J.M.; Nowicki, T.S.; Tsoi, J.; Puig-Saus, C.; Baselga-Carretero, I.; Medina, E.; Quist, M.J.; et al. PAK4 inhibition improves PD-1 blockade immunotherapy. Nat. Cancer 2020, 1, 46–58. [Google Scholar] [CrossRef] [Green Version]
  210. Sumimoto, H.; Imabayashi, F.; Iwata, T.; Kawakami, Y. The BRAF-MAPK signaling pathway is essential for cancer-immune evasion in human melanoma cells. J. Exp. Med. 2006, 203, 1651–1656. [Google Scholar] [CrossRef] [Green Version]
  211. Peng, D.; Kryczek, I.; Nagarsheth, N.; Zhao, L.; Wei, S.; Wang, W.; Sun, Y.; Zhao, E.; Vatan, L.; Szeliga, W.; et al. Epigenetic silencing of TH1-type chemokines shapes tumour immunity and immunotherapy. Nature 2015, 527, 249–253. [Google Scholar] [CrossRef] [PubMed]
  212. Nagarsheth, N.; Peng, D.; Kryczek, I.; Wu, K.; Li, W.; Zhao, E.; Zhao, L.; Wei, S.; Frankel, T.; Vatan, L.; et al. PRC2 Epigenetically Silences Th1-Type Chemokines to Suppress Effector T-Cell Trafficking in Colon Cancer. Cancer Res. 2016, 76, 275–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Topper, M.J.; Vaz, M.; Chiappinelli, K.B.; DeStefano Shields, C.E.; Niknafs, N.; Yen, R.C.; Wenzel, A.; Hicks, J.; Ballew, M.; Stone, M.; et al. Epigenetic Therapy Ties MYC Depletion to Reversing Immune Evasion and Treating Lung Cancer. Cell 2017, 171, 1284–1300.e21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Tauriello, D.V.F.; Palomo-Ponce, S.; Stork, D.; Berenguer-Llergo, A.; Badia-Ramentol, J.; Iglesias, M.; Sevillano, M.; Ibiza, S.; Cañellas, A.; Hernando-Momblona, X.; et al. TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 2018, 554, 538–543. [Google Scholar] [CrossRef] [Green Version]
  215. Vanpouille-Box, C.; Diamond, J.M.; Pilones, K.A.; Zavadil, J.; Babb, J.S.; Formenti, S.C.; Barcellos-Hoff, M.H.; Demaria, S. TGFβ Is a Master Regulator of Radiation Therapy-Induced Antitumor Immunity. Cancer Res. 2015, 75, 2232–2242. [Google Scholar] [CrossRef] [Green Version]
  216. Feig, C.; Jones, J.O.; Kraman, M.; Wells, R.J.; Deonarine, A.; Chan, D.S.; Connell, C.M.; Roberts, E.W.; Zhao, Q.; Caballero, O.L.; et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc. Natl. Acad. Sci. USA 2013, 110, 20212–20217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Bockorny, B.; Semenisty, V.; Macarulla, T.; Borazanci, E.; Wolpin, B.M.; Stemmer, S.M.; Golan, T.; Geva, R.; Borad, M.J.; Pedersen, K.S.; et al. BL-8040, a CXCR4 antagonist, in combination with pembrolizumab and chemotherapy for pancreatic cancer: The COMBAT trial. Nat. Med. 2020, 26, 878–885. [Google Scholar] [CrossRef]
  218. Kalbasi, A.; Ribas, A. Tumour-intrinsic resistance to immune checkpoint blockade. Nat. Rev. Immunol. 2020, 20, 25–39. [Google Scholar] [CrossRef]
  219. Wei, S.C.; Anang, N.A.S.; Sharma, R.; Andrews, M.C.; Reuben, A.; Levine, J.H.; Cogdill, A.P.; Mancuso, J.J.; Wargo, J.A.; Pe’er, D.; et al. Combination anti-CTLA-4 plus anti-PD-1 checkpoint blockade utilizes cellular mechanisms partially distinct from monotherapies. Proc. Natl. Acad. Sci. USA 2019, 116, 22699–22709. [Google Scholar] [CrossRef] [Green Version]
  220. Vanpouille-Box, C.; Hoffmann, J.A.; Galluzzi, L. Pharmacological modulation of nucleic acid sensors - therapeutic potential and persisting obstacles. Nat. Rev. Drug Discov. 2019, 18, 845–867. [Google Scholar] [CrossRef]
  221. Li, T.; Chen, Z.J. The cGAS-cGAMP-STING pathway connects DNA damage to inflammation, senescence, and cancer. J. Exp. Med. 2018, 215, 1287–1299. [Google Scholar] [CrossRef] [PubMed]
  222. Grasso, C.S.; Giannakis, M.; Wells, D.K.; Hamada, T.; Mu, X.J.; Quist, M.; Nowak, J.A.; Nishihara, R.; Qian, Z.R.; Inamura, K.; et al. Genetic Mechanisms of Immune Evasion in Colorectal Cancer. Cancer Discov. 2018, 8, 730–749. [Google Scholar] [CrossRef] [Green Version]
  223. Spranger, S.; Gajewski, T.F. Impact of oncogenic pathways on evasion of antitumour immune responses. Nat. Rev. Cancer 2018, 18, 139–147. [Google Scholar] [CrossRef] [PubMed]
  224. Wang, H.; Franco, F.; Tsui, Y.C.; Xie, X.; Trefny, M.P.; Zappasodi, R.; Mohmood, S.R.; Fernández-García, J.; Tsai, C.H.; Schulze, I.; et al. CD36-mediated metabolic adaptation supports regulatory T cell survival and function in tumors. Nat. Immunol. 2020, 21, 298–308. [Google Scholar] [CrossRef]
  225. Landry, L.G.; Ali, N.; Williams, D.R.; Rehm, H.L.; Bonham, V.L. Lack Of Diversity In Genomic Databases Is A Barrier To Translating Precision Medicine Research Into Practice. Health Aff. (Millwood) 2018, 37, 780–785. [Google Scholar] [CrossRef] [PubMed]
  226. Berger, T.R.; Wen, P.Y.; Lang-Orsini, M.; Chukwueke, U.N. World Health Organization 2021 Classification of Central Nervous System Tumors and Implications for Therapy for Adult-Type Gliomas: A Review. JAMA Oncol. 2022, 8, 1493–1501. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Immune cell and cancer cell or antigen-presenting cell receptor-ligand interactions involved in immune checkpoint modulation. The figure illustrates an overview of implicated receptor-ligand interactions and their general effects on the immune response. Several immune cells, such as CD8 + T cells, CD4 + T cells, NK cells, and Tregs, express specific receptors, which are recognized and bound by their specific ligands present on the surface of various cancer cells or antigen-presenting cells. The illustration also depicts examples of different receptors and ligands involved in ICI modulation, along with generalized stimulatory (↑) or inhibitory (↓) effects. ICIs block various inhibitory receptor-ligand interactions leading to the activation of immune cells, which leads to tumor regression. ICI therapies, including anti-PD-1/PD-L1 and anti-CTLA-4, have shown clinical efficacy for many cancers, which provided opportunities for developing alternative ICIs (e.g., anti-LAG-3, anti-TIM-3, and anti-NKG2). Adapted from ref. [3,34]. Created with BioRender.com (accessed on 18 November 2022). Definitions of abbreviations are found in the abbreviations list at the end of the text.
Figure 1. Immune cell and cancer cell or antigen-presenting cell receptor-ligand interactions involved in immune checkpoint modulation. The figure illustrates an overview of implicated receptor-ligand interactions and their general effects on the immune response. Several immune cells, such as CD8 + T cells, CD4 + T cells, NK cells, and Tregs, express specific receptors, which are recognized and bound by their specific ligands present on the surface of various cancer cells or antigen-presenting cells. The illustration also depicts examples of different receptors and ligands involved in ICI modulation, along with generalized stimulatory (↑) or inhibitory (↓) effects. ICIs block various inhibitory receptor-ligand interactions leading to the activation of immune cells, which leads to tumor regression. ICI therapies, including anti-PD-1/PD-L1 and anti-CTLA-4, have shown clinical efficacy for many cancers, which provided opportunities for developing alternative ICIs (e.g., anti-LAG-3, anti-TIM-3, and anti-NKG2). Adapted from ref. [3,34]. Created with BioRender.com (accessed on 18 November 2022). Definitions of abbreviations are found in the abbreviations list at the end of the text.
Ijms 24 00041 g001
Figure 2. Mechanisms of distinct tumor phenotypes, including immune-inflamed, immune-desert, and immune-excluded tumors, are associated with specific inhibitory and stimulatory biological mechanisms. Immune-inflamed tumors are permissive to immune cell infiltration; however, immune cells in the TME can be suppressed due to checkpoint activation. On the other hand, immune-desert tumors may be devoid of T cell priming due to the lack of tumor antigens, defective antigen processing and presentation processes, or impaired interactions between dendritic cells and T cells. The immune-excluded tumors, on the other hand, may display aberrant chemokines, activated oncogenic pathways, hypoxia, aberrant vasculature, or an immunosuppressive TME (e.g., stromal barriers). Adapted from ref. [124]. ↓ (downward arrow), inhibitors; ↑ (upward arrow), stimulatory factors. Created with BioRender.com (URL accessed on 18 November 2022). Definitions of abbreviations are found in the abbreviations list at the end of the text.
Figure 2. Mechanisms of distinct tumor phenotypes, including immune-inflamed, immune-desert, and immune-excluded tumors, are associated with specific inhibitory and stimulatory biological mechanisms. Immune-inflamed tumors are permissive to immune cell infiltration; however, immune cells in the TME can be suppressed due to checkpoint activation. On the other hand, immune-desert tumors may be devoid of T cell priming due to the lack of tumor antigens, defective antigen processing and presentation processes, or impaired interactions between dendritic cells and T cells. The immune-excluded tumors, on the other hand, may display aberrant chemokines, activated oncogenic pathways, hypoxia, aberrant vasculature, or an immunosuppressive TME (e.g., stromal barriers). Adapted from ref. [124]. ↓ (downward arrow), inhibitors; ↑ (upward arrow), stimulatory factors. Created with BioRender.com (URL accessed on 18 November 2022). Definitions of abbreviations are found in the abbreviations list at the end of the text.
Ijms 24 00041 g002
Figure 3. Stimulatory and inhibitory factors in the cancer-immunity cycle. Each step of the cancer-immunity cycle requires the coordination of several stimulatory and inhibitory factors. Stimulatory factors shown in green with the upward arrow (↑) indicating promotion of immunity. Inhibitors shown in red with the downward arrow (↓) help keep the process in check and reduce immune activity and/or prevent autoimmunity. Examples of such factors and the primary steps at which they can act are shown. Antitumor immunity is mediated predominantly by CD8+ T cells and tumor immunity involves: (1) tumor antigen release, (2) tumor antigen processing and presentation by APCs (e.g., dendritic cells), (3) priming and activation of T cells, (4) trafficking of T cells via the bloodstream to tumors, (5) infiltration of activated T cells into the tumor parenchyma from the vasculature or tumor periphery, (6) recognition of tumor cells through antigenic peptide-MHC complexes on the surface of tumor cells by T cells, and (7) killing of tumor cells by cytotoxic T cells through granule exocytosis and release of perforin and granzyme or through the Fas/FasL pathway by inducing ferroptosis and pyroptosis. The release of additional antigens from dead tumor cells allows the continuation of the tumor-immunity cycle. Tumors with the immune-desert phenotype cannot pass steps 1–3 due to the absence of T cells in both the tumor and its margins. On the other hand, tumors with the immune-excluded phenotype cannot pass steps 4–5 due to the absence of T cells in the tumor bed. On the other hand, tumors with the immune-inflamed phenotype cannot pass steps 6–7 because of T cell exhaustion and checkpoint upregulation. Immune checkpoint proteins, such as PD1 and CTLA-4, can suppress the development of an active immune response by acting primarily at the level of T cell development and proliferation (step 3). Adapted from ref. [124,125]. Created with BioRender.com (URL (accessed on 18 November 2022). Definitions of abbreviations are found in the abbreviations list at the end of the text.
Figure 3. Stimulatory and inhibitory factors in the cancer-immunity cycle. Each step of the cancer-immunity cycle requires the coordination of several stimulatory and inhibitory factors. Stimulatory factors shown in green with the upward arrow (↑) indicating promotion of immunity. Inhibitors shown in red with the downward arrow (↓) help keep the process in check and reduce immune activity and/or prevent autoimmunity. Examples of such factors and the primary steps at which they can act are shown. Antitumor immunity is mediated predominantly by CD8+ T cells and tumor immunity involves: (1) tumor antigen release, (2) tumor antigen processing and presentation by APCs (e.g., dendritic cells), (3) priming and activation of T cells, (4) trafficking of T cells via the bloodstream to tumors, (5) infiltration of activated T cells into the tumor parenchyma from the vasculature or tumor periphery, (6) recognition of tumor cells through antigenic peptide-MHC complexes on the surface of tumor cells by T cells, and (7) killing of tumor cells by cytotoxic T cells through granule exocytosis and release of perforin and granzyme or through the Fas/FasL pathway by inducing ferroptosis and pyroptosis. The release of additional antigens from dead tumor cells allows the continuation of the tumor-immunity cycle. Tumors with the immune-desert phenotype cannot pass steps 1–3 due to the absence of T cells in both the tumor and its margins. On the other hand, tumors with the immune-excluded phenotype cannot pass steps 4–5 due to the absence of T cells in the tumor bed. On the other hand, tumors with the immune-inflamed phenotype cannot pass steps 6–7 because of T cell exhaustion and checkpoint upregulation. Immune checkpoint proteins, such as PD1 and CTLA-4, can suppress the development of an active immune response by acting primarily at the level of T cell development and proliferation (step 3). Adapted from ref. [124,125]. Created with BioRender.com (URL (accessed on 18 November 2022). Definitions of abbreviations are found in the abbreviations list at the end of the text.
Ijms 24 00041 g003
Figure 4. Mechanisms of resistance to immunotherapy. (A) Intrinsic mechanisms of resistance to immunotherapy. Examples of intrinsic mechanisms of adaptive resistance involve altered signaling pathways, limited mutational burden, de-differentiation of tumor resulting in loss of neoantigen expression, defective antigen processing, constitutive PD-L1 expression, and loss of HLA expression. Examples of intrinsic mechanisms of acquired resistance include loss of antigenic target, loss of HLA expression, and escape mutations in IFN signaling. (B) Extrinsic mechanisms of resistance to ICI therapy (right panel). Examples of extrinsic mechanisms of resistance involve upregulated or constitutive immune checkpoint expression, immunosuppressive cytokine release (e.g., CSF-1, TGFβ, adenosine) within the TME, T cell exhaustion and phenotypic switching, as well as elevation of immunosuppressive cell populations (e.g., Treg, MDSC, and M2 macrophages). Adapted from ref. [34]. Created with BioRender.com (URL (accessed on 18 November 2022). Definitions of abbreviations are found in the abbreviations list at the end of the text.
Figure 4. Mechanisms of resistance to immunotherapy. (A) Intrinsic mechanisms of resistance to immunotherapy. Examples of intrinsic mechanisms of adaptive resistance involve altered signaling pathways, limited mutational burden, de-differentiation of tumor resulting in loss of neoantigen expression, defective antigen processing, constitutive PD-L1 expression, and loss of HLA expression. Examples of intrinsic mechanisms of acquired resistance include loss of antigenic target, loss of HLA expression, and escape mutations in IFN signaling. (B) Extrinsic mechanisms of resistance to ICI therapy (right panel). Examples of extrinsic mechanisms of resistance involve upregulated or constitutive immune checkpoint expression, immunosuppressive cytokine release (e.g., CSF-1, TGFβ, adenosine) within the TME, T cell exhaustion and phenotypic switching, as well as elevation of immunosuppressive cell populations (e.g., Treg, MDSC, and M2 macrophages). Adapted from ref. [34]. Created with BioRender.com (URL (accessed on 18 November 2022). Definitions of abbreviations are found in the abbreviations list at the end of the text.
Ijms 24 00041 g004
Figure 5. Genomic correlations with response and resistance to ICI therapy within the immune TME. The panel (A) represents correlations with response, focusing on antigen presentation and recognition. The panel (B) represents resistance pathways that promote tumor immune evasion mechanisms that induce immunosuppressive cells, leading to the inhibition of T cell-mediated anti-tumor response (right side). Adapted from [113]. The panel (B) also illustrates the TME, which consists of cellular and non-cellular components. The cellular component consists of cancer cells, endothelial cells, pericytes, carcinoma-associated fibroblasts, and immune cells. The immune compartment comprises many immune cell populations (e.g., T cells, B cells, natural killer cells, tumor-associated macrophages, myeloid-derived suppressor cells, and dendritic cells). The non-cellular component of the TME, on the other hand, is represented by the extracellular matrix and functions as a scaffold. Components of the TME interact via the extracellular matrix, cell-cell contacts, and through the release of cytokines, chemokines, extracellular vesicles, and others. Adapted from ref. [113,184,185]. Definitions of abbreviations are found in the abbreviations list at the end of the text.
Figure 5. Genomic correlations with response and resistance to ICI therapy within the immune TME. The panel (A) represents correlations with response, focusing on antigen presentation and recognition. The panel (B) represents resistance pathways that promote tumor immune evasion mechanisms that induce immunosuppressive cells, leading to the inhibition of T cell-mediated anti-tumor response (right side). Adapted from [113]. The panel (B) also illustrates the TME, which consists of cellular and non-cellular components. The cellular component consists of cancer cells, endothelial cells, pericytes, carcinoma-associated fibroblasts, and immune cells. The immune compartment comprises many immune cell populations (e.g., T cells, B cells, natural killer cells, tumor-associated macrophages, myeloid-derived suppressor cells, and dendritic cells). The non-cellular component of the TME, on the other hand, is represented by the extracellular matrix and functions as a scaffold. Components of the TME interact via the extracellular matrix, cell-cell contacts, and through the release of cytokines, chemokines, extracellular vesicles, and others. Adapted from ref. [113,184,185]. Definitions of abbreviations are found in the abbreviations list at the end of the text.
Ijms 24 00041 g005
Figure 6. Approaches to turn a “cold tumor” into a “hot tumor”. Some representative therapeutic strategies to increase T cell infiltration and improve the efficacy of ICI. T cell priming and activation: Oncolytic viruses, local thermal ablation therapy (e.g., radiofrequency ablation), chemotherapy, and radiotherapy are all capable of inducing immunogenic cell death to promote T cell priming and activation. Local administration of immune adjuvants, such as TLR agonists, promotes the activation of DCs. Epigenetic modification inhibitors can promote T cell priming by increasing the expression of tumor antigens and by restoring antigen processing and presentation mechanisms. T cell expansion: Cancer vaccines and adoptive cellular therapies, such as CAR-T cells, can promote the expansion of tumor-specific T cells. T cell trafficking and infiltration: Intrinsic oncogenic pathway inhibitors, antiangiogenic therapies, TGFβ inhibitors, CXCR4 inhibitors, and epigenetic modification inhibitors can promote T cell trafficking and enable T cells to infiltrate the tumor more effectively. Adapted from ref. [124]. Created with Briorender.com (URL (accessed on 18 November 2022). Definitions of abbreviations are found in the abbreviations list at the end of the text.
Figure 6. Approaches to turn a “cold tumor” into a “hot tumor”. Some representative therapeutic strategies to increase T cell infiltration and improve the efficacy of ICI. T cell priming and activation: Oncolytic viruses, local thermal ablation therapy (e.g., radiofrequency ablation), chemotherapy, and radiotherapy are all capable of inducing immunogenic cell death to promote T cell priming and activation. Local administration of immune adjuvants, such as TLR agonists, promotes the activation of DCs. Epigenetic modification inhibitors can promote T cell priming by increasing the expression of tumor antigens and by restoring antigen processing and presentation mechanisms. T cell expansion: Cancer vaccines and adoptive cellular therapies, such as CAR-T cells, can promote the expansion of tumor-specific T cells. T cell trafficking and infiltration: Intrinsic oncogenic pathway inhibitors, antiangiogenic therapies, TGFβ inhibitors, CXCR4 inhibitors, and epigenetic modification inhibitors can promote T cell trafficking and enable T cells to infiltrate the tumor more effectively. Adapted from ref. [124]. Created with Briorender.com (URL (accessed on 18 November 2022). Definitions of abbreviations are found in the abbreviations list at the end of the text.
Ijms 24 00041 g006
Table 1. A general representation of predictive biomarkers of response to immune checkpoint inhibitors.
Table 1. A general representation of predictive biomarkers of response to immune checkpoint inhibitors.
Tumor CellsTumor MicroenvironmentCirculating FactorsHost FactorsImmune-Related
Adverse Events
PDL-1 expressionPDL-1 expressionPeripheral blood cells, e.g., myelogenous cells, eosinophils, macrophages, CD+ ICSO+ T cellsAge, gender, body fat distributionEndocrine immune-related adverse events, e.g., thyroid dysfunction Skin immune-related adverse events e.g., vitiligo, pruritus, lichenoid toxicity
TMBTumor-infiltrating lymphocytes, e.g., CD39+ CD8+ T cells. CD4+ T cells, FOXP3+ T cells, TAMs, myeloid cells, NKp46+ cellsOther circulating factors, e.g., PDL-1, soluble proteins, cytokines and inflammatory factorsHost germline mutations, HLA diversity, and other specific mutations
DDR pathways: dMMR/MSIImmune status of tumor microenvironmentCirculating nucleic acids, e.g., ctDNA, RNA (mRNA, miRNA)Intestinal commensal microbiota
Specific gene mutationsImmunologic classification, immunoscoreCirculating tumor cells (CTCs)
Neoantigen loadDiversity of immune cell repertoire
TIL richness and clonality
Predictive biomarkers of response to ICI therapies include the expression of intermolecular interactions within tumor cells to the expression of various molecules and cells in the TME and also circulating tumor and host factors. Adapted from ref. [36]. Definitions of abbreviations are found in the abbreviations list at the end of the text.
Table 2. Types of biomarkers and their clinical utility in metastatic melanoma.
Table 2. Types of biomarkers and their clinical utility in metastatic melanoma.
Diagnostic BiomarkersPrognostic BiomarkersPredictive BiomarkersOn-Treatment Biomarkers
PurposePresence of diseaseOverall patient survival independent of therapyResponse to treatment (efficacy or toxicity)Pharmacodynamic biomarkers (drug interaction with its target)
Time of
evaluation
Before diagnosis and at diagnosisAt diagnosisBefore treatment
selection
During or post-treatment
Clinical utilityBefore diagnosis: Allows screening of healthy patients
At diagnosis: Stratifies benign vs. malignant, classify into subtypes
At diagnosis: Estimate risk of disease
Post diagnosis: Allows monitoring disease status, detects recurrence
Identify treatments likely to be effective, guides initial treatment strategy and decision makingDetermines degree of drug response, guides treatments decision making during treatment
Current state Validated Emerging
Human Melanoma Black-45 (HMB-45)Lactate dehydrogenaseBRAF V600 mutationTMBctDNA profiles
Melan-AM stage Neoantigen loadAbsolute lymphocyte count
TyrosinaseDisease sites Molecular alterationsProliferating CD8+ T cells
Microphthalmia transcription factor (MITF) PDL-1 expressionIncrease of T-cell subsets and checkpoint molecules (PDL-1, LAG-3)
S100 LAG-3 expressionGranzyme B expression
SM5-1 CD8+ T cells at tumor invasive marginT-cell receptor (TCR) signature
Chondroitin sulfate proteoglycan 4 (CSPG4) IL-17 expression
Loss of p16 protein expression Immune-related gene expression signatures
Biomarker panels and gene arrays T-cell receptor (TCR) signature
Adapted from ref. [37,38,39,40]. Definitions of abbreviations are found in the abbreviations list at the end of the text.
Table 3. Genomic correlations with response and resistance based on primary location.
Table 3. Genomic correlations with response and resistance based on primary location.
Primary LocationResponse CategoryCharacteristics or ModalityReferences
T cellIntratumoral infiltrationTranscriptional signatures of cytotoxic lymphocytes infiltrating the tumor core[67,83,141,142,143,144]
Enhanced effector functionIncreased expression of PRF1, GZMA/B, CD8A, and IFNG[82,95,145]
Increased clonalityRanging from 0 to 1, with 1 indicating a monoclonal population[46,121,146]
Greater stemnessExpress chemokine receptor CXCR5 and transcription factor TCF7; lack TIM-3/CD39[98,147]
Reduced exhaustionExpress co-inhibitory receptor TIM-3 and ectonucleotidase CD39; lack CXCR5/TCF7[98,147]
Tumor cell
(response
mechanisms)
Tumor antigensNeoantigens, viral antigens[7,66,69,72,131,148,149,150,151,152,153,154,155]
Increased tumor mutation burdenMismatch repair deficiency[151,156,157]
Immunogenic alterationsInactivating mutations in SERPINB3 and SERPINB4[158]
Mutational signaturesSmoking, ultraviolet light, alkylating agent therapy, APOBEC[66,159,160]
Genomic upregulation of PD-L1PDL1 amplification and loss of CDK4, SPOP, and CMTM4 and CMTM6[130,161,162,163,164,165,166,167]
Chromatin modifier lossInactivating mutations in PBRM1, ARID1A, and SMARCA4[159,168,169,170]
Tumor cell
(resistance
mechanisms)
Tumor antigensCancer/testis antigens similar to self and less immunogenic[171]
Deficient antigen presentationInactivating mutations in B2M, HLA, JAK/STAT, and IFN-γ response genes[151,159]
Oncogenic pathwaysInactivating STK11 and PTEN mutations, WNT/β-catenin, EGFR and KRAS mutations[67,135,172,173,174,175,176,177,178,179]
Immune evasion alterationsIncreased expression of SERPINB9[82]
CNAsHigh levels of copy-number loss, chromosome arm and whole-chromosome CNAs[46,73]
Tumor
microenvironment
Immunosuppressive stromal cellsTranscriptional signatures of fibroblasts, endothelial cells, and TGF-β signaling[30,67,180,181]
Immunosuppressive immune cellsTranscriptional signatures of myeloid-derived suppressor cells and regulatory T cells[82,142]
Adapted from ref. [113]. Definitions of abbreviations are found in the abbreviations list at the end of the text.
Table 4. Therapeutic strategies to turn “cold tumors” into “hot tumors”.
Table 4. Therapeutic strategies to turn “cold tumors” into “hot tumors”.
Therapeutic ModalitiesPrimary Effect SiteMain MechanismsReferences
Immune adjuvants (TLR agonists, STING agonists)Draining lymph nodePromotes T cell priming by antigen release, antigen processing and presentation, and DC-T cell interaction[187,188]
Oncolytic viruses[189,190]
Chemotherapy and radiotherapy[191,192]
Epigenetic modification inhibitors (DNMT inhibitor, HDAC inhibitor, EZH2 inhibitor)[193,194,195]
Metabolic intervention[196,197]
Local thermal ablation therapy (Radiofrequency ablation)[198]
Photothermal therapy and photodynamic therapy[26,199,200]
Magnetic hyperthermia[201,202]
High-intensity focused ultrasound[203,204]
Adoptive cellular therapy (TILs, CAR-T cells)Draining lymph nodePromotes T cell expansion[205,206,207]
Vaccines[208]
Oncogenic pathway inhibitorsTumor microenvironmentPromotes T cell trafficking and infiltration[174,209,210]
Epigenetic modification inhibitors[211,212,213]
Antiangiogenic therapy (anti-VEGF)[102]
TGFβ inhibitors[180,214,215]
CXCR4 inhibitors[216,217]
Examples of therapeutic approaches to drive T cells into tumors include TLR and STING agonists [187,188], oncolytic viruses [189,190], chemotherapy and radiotherapy [191,192]], epigenetic modification inhibitors (DNMT inhibitor, HDAC inhibitor, EZH2 inhibitor) [193,194,195], metabolic intervention [196,197], local thermal ablation therapy (radiofrequency ablation) [198], photothermal therapy and photodynamic therapy [26,199,200], magnetic hyperthermia [201,202], and high-intensity focused ultrasound [203,204], which all promote T cell priming by antigen release, antigen processing and presentation, and DC-T cell interaction [124]. Meanwhile, adoptive cellular therapy (TILs, CAR-T cells) [205,206,207] and vaccines [208] promote T cell expansion [124]. On the other hand, oncogenic pathway inhibitors [174,209,210], epigenetic modification inhibitors [211,212,213], antiangiogenic therapy (anti-VEGF) [102], TGFβ inhibitors [180,214,215], and CXCR4 inhibitors promote T cell trafficking and infiltration [216,217]. Adapted from ref. [124]. Definitions of abbreviations are found in the abbreviations list at the end of the text.
Table 5. Representative examples of ICI therapy in combination with other drugs to overcome resistance.
Table 5. Representative examples of ICI therapy in combination with other drugs to overcome resistance.
TitleInterventionsTrial NoConditionsPhaseSettingStatus as of
11-2022
Neoantigen vaccines
A Phase I Study Combining NeoVax, a Personalized NeoAntigen Cancer Vaccine, With Ipilimumab to Treat High-risk Renal Cell CarcinomaNeoVax  +  anti-CTLA-4 ipilimumabNCT02950766Stage III or low-volume stage IV renal cell
carcinoma
IAdjuvant/1st lineRecruiting
Neoantigen-based Personalized Vaccine Combined with Immune Checkpoint Blockade Therapy in Patients with Newly Diagnosed, Unmethylated GlioblastomaNeoVax  +  anti-PD-1 nivolumab  ±  anti-CTLA-4 ipilimumabNCT03422094Newly diagnosed, unmethylated glioblastomaI1st lineTerminated (Manufacturer changed focus to cell therapy)
Personalized Neoantigen Cancer Vaccine + Pembrolizumab After Rituximab for Follicular LymphomaNeoVax  +  anti-CD20 rituxumabNCT03361852Follicular lymphomaI1st lineRecruiting
A Personalized Neoantigen Cancer Vaccine in Treatment Naïve, Asymptomatic Patients with IGHV Unmutated CLLNeoVax  +  alkylating agent cyclophosphamideNCT03219450Chronic lymphocytic leukemiaI1st lineRecruiting
Pooled Mutant KRAS-Targeted Long Peptide Vaccine Combined with Nivolumab and Ipilimumab for Patients with Resected MMR-p Colorectal and Pancreatic CancerKRAS peptide vaccine, nivolumab, ipilimumabNCT04117087Colorectal cancer, pancreatic cancerIRecurrent or metastaticRecruiting
NeoVax Plus Ipilimumab in Renal Cell CarcinomaNeoVax, ipilimumabNCT02950766Kidney cancerI1st lineRecruiting
Immunotherapy Combined with Radiation and Influenza Vaccine for Pancreatic CancerNivolumab, ipilimumab, influenza vaccine, stereotactic body radiation therapyNCT05116917Pancreatic
cancer
IIRecurrentRecruiting
Combination Therapy with Nivolumab and PD-L1/IDO Peptide Vaccine to Patients with Metastatic MelanomaNivolumab, PD-L1/IDO peptide vaccineNCT03047928Metastatic melanomaI/IIRecurrentRecruiting
A Phase I/II Trial to Evaluate a Peptide Vaccine Plus Ipilimumab in Patients with MelanomaIpilimumab, peptide vaccine 6MHPNCT02385669MelanomaI/IIStage III or IV, recurrent or metastatic, stage IIA, IIB-IV resected to no evidence of diseaseTerminated, has Results
Vaccine Combining Multiple Class I Peptides and Montanide ISA 51VG With Escalating Doses of Anti-PD-1 Antibody Nivolumab or Ipilimumab with Nivolumab For Patients with Resected Stages IIIC/IV MelanomaNY-ESO-1 157-165 (165 V), nivolumab, gp100:280-288 (288 V), montanide ISA 51 vegetable grade (VG), ipilimumabNCT01176474MelanomaIStages IIIC/IV melanoma, with no evidence of diseaseActive, not recruiting
Dendritic cell vaccines
Combination Immunotherapy-Ipilimumab-Nivolumab-Dendritic Cell p53 Vac - Patients with Small Cell Lung CancerDendritic cell-based p53 vaccine + nivolumab + ipilimumabNCT03406715Small cell lung cancerIIRecurrentActive, not recruiting
Nivolumab with DC Vaccines for
Recurrent Brain
Tumors
Dentritic cell + nivolumabNCT02529072Malignant glioma, astrocytoma, glioblastomaIRecurrentCompleted, has results
Neoantigen Dendritic Cell Vaccine and Nivolumab in HCC and Liver Metastases from CRCNeoantigen dendritic cell vaccine + nivolumabNCT03782064Hepatocellular carcinoma, hepatocellular cancer, colorectal cancer, colorectal carcinoma, liver metastasesIINewly diagnosed or recurrent hepatocellular carcinomaRecruiting
Dendritic Cell (DC)/Myeloma Fusions in Combination with Nivolumab in Patients with Relapsed Multiple Myeloma NCT03782064Multiple myelomaIIRelapsedTerminated
Has Results
Polarized Dendritic Cell (aDC1) Vaccine, Interferon Alpha-2, Rintatolimod, and Celecoxib for the Treatment of HLA-A2 + Refractory MelanomaAlpha-type-1 polarized dendritic cells, celecoxib, PD-L1 inhibitor, PD-1 inhibitor, recombinant Interferon alpha-2b, rintatolimodNCT04093323HLA-A2 positive cells present, refractory melanomaIIRefractoryRecruiting
Interferon therapies
Testing the Combination of Two Experimental Drugs MK-3475 (Pembrolizumab) and Interferon-gamma for the Treatment of Mycosis Fungoides and Sézary Syndrome and Advanced Synovial SarcomaInterferon-γ-1β + anti-PD-1 pembrolizumabNCT03063632Mycosis fungoides and Sézary syndromeIIRefractoryActive, not recruiting
Combination of Interferon-gamma and Nivolumab for Advanced Solid TumorsInterferon-γ  +  anti-PD-1 nivolumabNCT02614456Advanced solid tumorsI2nd lineCompleted
Pembrolizumab Combined with Itacitinib (INCB039110) and/or Pembrolizumab Combined with INCB050465 in Advanced Solid TumorsJAK1 inhibitor itacitinib or PI3Kδ inhibitor  +  anti-PD-1 pembrolizumabNCT02646748Advanced solid tumorsI/IIRefractoryCompleted
Pembrolizumab and Ruxolitinib Phosphate in Treating Patients with Metastatic Stage IV Triple Negative Breast CancerJAK2 inhibitor ruxolitinib  +  anti-PD-1 pembrolizumabNCT03012230Metastatic triple-negative breast cancerI2nd lineRecruiting
Pembrolizumab and Itacitinib (INCB039110) for Non-Small Cell Lung CancerJAK1 inhibitor itacitinib  +  anti-PD-1 pembrolizumabNCT03425006Metastatic PD-L1+  non-small cell lung cancerII1st lineTerminated
Study of the Safety and Efficacy of MIW815 with PDR001 in Patients with Advanced/Metastatic Solid Tumors or LymphomasSTING agonist MIW815  +  anti-PD-1 spartalizumabNCT03172936Advanced solid tumors or lymphomasIAny lineTerminated (Sponsor’s decision)
Trial of Intratumoral Injections of TTI-621 in Subjects with Relapsed and Refractory Solid Tumors and Mycosis
Fungoides
TTI-621 monotherapy,
TTI-621 + PD-1/PD-L1 inhibitor, TTI-621 + pegylated interferon-α2aOther: TTI-621 + T-Vec, TTI-621 + radiation
TTI-621 (SIRPα-IgG1 Fc) anti- CD47 “don’t eat me” signal
NCT02890368Solid tumors, mycosis fungoides, melanoma, merkel-cell carcinoma, squamous cell carcinoma, breast carcinoma, human papillomavirus-related malignant neoplasm, soft tissue sarcomaIRelapsed and refractoryTerminated
A Study to Assess the Safety and Tolerability of Atezolizumab in Combination with Other Immune-Modulating Therapies in Participants with Locally Advanced or Metastatic Solid TumorsAtezolizumab, bevacizumab, interferon alpha-2b, ipilimumab, obinutuzumab, PEG-interferon alpha-2aNCT02174172Solid tumorsILocally advanced or metastatic solid tumorsCompleted
The Study of JS001 Compared to High-Dose Interferon in Patients with Mucosal Melanoma That Has Been Removed by SurgeryHumanized anti-PD-1 monoclonal antibody toripalimab, high-dose recombinant interferon a-2BNCT03178123Mucosal melanomaII1st line after surgeryActive, not
recruiting
Ipilimumab or High-Dose Interferon Alfa-2b in Treating Patients with High-Risk Stage III–IV Melanoma That Has Been Removed by SurgeryIpilimumab, interferon alpha-2bNCT01274338Melanoma of unknown primary, recurrent melanoma, stage IIIB cutaneous melanoma AJCC v7, stage IIIC cutaneous melanoma AJCC v7, stage IV cutaneous melanoma AJCC v6 and v7IIIRecurrence after surgeryActive, not recruiting, has results
Ipilimumab With or Without High-Dose Recombinant Interferon Alfa-2b in Treating Patients with Stage III–IV Melanoma That Cannot Be Removed by SurgeryIpilimumab, recombinant interferon alpha-2bNCT01708941Recurrent melanoma, stage IIIA cutaneous melanoma AJCC v7, stage IIIB cutaneous melanoma AJCC v7, stage IIIC cutaneous melanoma AJCC v7, stage IV cutaneous melanoma AJCC v6 and v7IIUnresectable stage III or stage IV melanoma, either initial presentation or recurrentActive, not recruiting, has results
Nivolumab, Fluorouracil, and Interferon Alpha 2B for the Treatment of Unresectable Fibrolamellar CancerFluorouracil, nivolumab, recombinant interferon alpha 2b-like proteinNCT04380545Stage III IVB epatocellular carcinoma AJCC v8, unresectable fibrolamellar carcinomaI/II1st lineRecruiting
High-Dose Recombinant Interferon Alfa-2B, Ipilimumab, or Pembrolizumab in Treating Patients with Stage III–IV High Risk Melanoma That Has Been Removed by SurgeryIpilimumab, pembrolizumab, recombinant interferon alpha-2bNCT02506153Cutaneous melanoma, metastatic mucosal melanoma, metastatic non-cutaneous melanoma, non-cutaneous melanoma, recurrent cutaneous melanoma, recurrent mucosal melanoma, recurrent non-cutaneous melanoma, and stage III-IVC of all aboveIII1st line, high risk melanoma that has been removed by surgeryActive, not recruiting
Safety and Tolerability of Pembrolizumab (MK-3475) + Pegylated Interferon Alfa-2b and Pembrolizumab+ Ipilimumab in Participants with Advanced Melanoma or Renal Cell Carcinoma (MK-3475-029/KEYNOTE-29)Pembrolizumab, PegIFN-2b, IpilimumabNCT02089685Renal cell carcinoma, melanomaI/IIAdvanced or unresectable or metastatic renal cell carcinoma, melanomaCompleted
Has Results
IPI-Biotherapy for Patients Previously Treated with Cytotoxic Drugs with Metastatic MelanomaIpilimumab, interferon, interleukin-2 (aldesleukin)NCT01409187MelanomaI/IIRecurrentWithdrawn
TIL and Anti-PD1 in Metastatic MelanomaNivolumab & tumor- infiltrating lymphocytes with/without interferon-alphaNCT03638375MelanomaI/IIRecurrentRecruiting
Trial of Intratumoral Injections of TTI-621 in Subjects with Relapsed and Refractory Solid Tumors and Mycosis FungoidesTTI-621 (SIRPα-IgG1 Fc) monotherapy
TTI-621 + PD-1/PD-L1 inhibitor, TTI-621 + pegylated interferon-α2a, TTI-621 + T-Vec, TTI-621 + radiation
NCT02890368Solid tumors, mycosis fungoides
Melanoma, merkel-cell carcinoma, squamous cell carcinoma, breast carcinoma, human papillomavirus-related malignant neoplasm,
soft tissue sarcoma
IRecurrentTerminated
CDK4/6 Inhibitors
Palbociclib After CDK and Endocrine Therapy (PACE)Fulvestrant  ±  CDK4/6 inhibitor palbociclib  ±  anti-PD-L1 avelumabNCT03147287Metastatic HR+HER2 breast cancerII2nd–3rd lineActive, not recruiting
Neoadjuvant Endocrine Therapy, Palbociclib, Avelumab in Estrogen Receptor Positive Breast Cancer (ImmunoADAPT)Tamoxifen  ±  CDK4/6 inhibitor palbociclib followed by anti-PD-L1 avelumabNCT03573648Stage II or III HR+ breast cancerIINeoadjuvantRecruiting
Ribociclib + PDR001 in Breast Cancer and Ovarian CancerCDK4/6 inhibitor ribociclib  +  anti-PD-1 spartalizumab ( + fulvestrant if breast)NCT03294694Metastatic ovarian cancer or HR+HER2 breast cancerIAny lineTerminated (safety implications)
A Study of Abemaciclib (LY2835219) in Participants with Non-Small Cell Lung Cancer or Breast CancerCDK4/6 inhibitor abemaciclib  +  anti-PD-1 pembrolizumabNCT02779751Metastatic non-small cell lung cancer or HR+HER2 breast cancerI1st–3rd lineActive, not
recruiting
Avelumab, Cetuximab, and Palbociclib in Recurrent or Metastatic Head and Neck Squamous Cell CarcinomaAnti-PD-L1 avelumab, cetuximab,  +  CDK4/6 inhibitor palbociclibNCT03498378Recurrent/metastatic head and neck squamous cell carcinomaIAny lineActive, not recruiting
Carboplatin, Etoposide, and Atezolizumab with or Without Trilaciclib (G1T28), a CDK4/6 Inhibitor, in Extensive-Stage SCLCAnti-PDL-1 atezolizumab, trilaciclib, carboplatin, etoposide, placeboNCT03041311Small cell lung cancerII1st lineTerminated, has results
Avelumab or Hydroxychloroquine with or Without Palbociclib to Eliminate Dormant Breast CancerAnti-PDL-1 avelumab, HCQ, palbociclibNCT04841148Breast cancerIIRefractoryRecruiting
Phase Ib Study of TNO155 in Combination with Spartalizumab or Ribociclib in Selected MalignanciesAnti-PD-1 spartalizumab,
ribociclib,
TNO155
NCT04000529Non-small cell lung cancer, head and neck squamous cell carcinoma, esophageal squamous cell carcinoma, gastrointestinal stromal tumors, colorectal cancerIRefractoryRecruiting
Ribociclib and Spartalizumab in R/M HNSCCAnti-PD-1 spartalizumab,
ribociclib,
NCT04213404Head and neck squamous cell carcinomaIRecurrentActive, not recruiting
Anti-TGF-β therapies
Phase I/Ib Study of NIS793 in Combination with PDR001 in Patients with Advanced MalignanciesAnti-TGF-β NIS793  +  anti-PD-1 spartalizumabNCT02947165Advanced solid tumorsIRefractoryCompleted
A Study of Galunisertib (LY2157299) and Durvalumab (MEDI4736) in Participants with Metastatic Pancreatic CancerTGF-β receptor 1 inhibitor galunisertib  +  anti-PD-L1 durvalumabNCT02734160Metastatic
pancreatic cancer
I3rd lineCompleted
A Study of Galunisertib (LY2157299) in Combination with Nivolumab in Advanced Refractory Solid Tumors and in Recurrent or Refractory NSCLC, or Hepatocellular CarcinomaTGF-β receptor 1 inhibitor galunisertib  +  anti-PD-1 nivolumabNCT02423343Recurrent or refractory non-small cell lung cancer or hepatocellular carcinomaI/II2nd lineCompleted
MSB0011359C (M7824) in Metastatic or Locally Advanced Solid TumorsAnti-PD-L1/TGF-β trap M7824NCT02517398Metastatic or locally advanced solid tumorsIRefractoryCompleted
M7824 in Treating Patients with Stage II–III HER2 Positive Breast CancerAnti-PD-L1/TGF-β trap M7824  +  treatment of physician’s choiceNCT03620201Stage II–III HER2+ breast cancerINeoadjuvantActive, not recruiting
M7824 vs. Pembrolizumab as a First line (1L) Treatment
in Participants with Programmed Death-ligand 1 (PD-L1) Expressing Advanced Non-small Cell Lung Cancer (NSCLC)
Anti-PD-L1/TGF-β trap M7824 vs. anti-PD-1 pembrolizumabNCT03631706PD-L1+  non-small cell lung cancerII1st lineActive, not recruiting
Antitumor Activity of Vactosertib in Combination with Pembrolizumab in Acral and Mucosal Melanoma Patients Progressed from Prior Immune Check Point InhibitorAnti-PD-1 pembrolizumab + anti-TGF-βR1 vactosertibNCT05436990Acral melanoma, mucosal melanomaIIRelapsedNot yet recruiting
Examples of recent clinical trials investigating various novel therapeutic modalities as interventions in combination with ICI to overcome monotherapy resistance and attempt to turn “cold tumors’ into “hot tumors”. Adapted from ref. [113]. Definitions of abbreviations are found in the abbreviations list at the end of the text.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Seyhan, A.A.; Carini, C. Insights and Strategies of Melanoma Immunotherapy: Predictive Biomarkers of Response and Resistance and Strategies to Improve Response Rates. Int. J. Mol. Sci. 2023, 24, 41. https://doi.org/10.3390/ijms24010041

AMA Style

Seyhan AA, Carini C. Insights and Strategies of Melanoma Immunotherapy: Predictive Biomarkers of Response and Resistance and Strategies to Improve Response Rates. International Journal of Molecular Sciences. 2023; 24(1):41. https://doi.org/10.3390/ijms24010041

Chicago/Turabian Style

Seyhan, Attila A., and Claudio Carini. 2023. "Insights and Strategies of Melanoma Immunotherapy: Predictive Biomarkers of Response and Resistance and Strategies to Improve Response Rates" International Journal of Molecular Sciences 24, no. 1: 41. https://doi.org/10.3390/ijms24010041

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