Next Article in Journal
The Human TOR Signaling Regulator Is the Key Indicator of Liver Cancer Patients’ Overall Survival: TIPRL/LC3/CD133/CD44 as Potential Biomarkers for Early Liver Cancers
Next Article in Special Issue
Nationwide Survival Benefit after Implementation of First-Line Immunotherapy for Patients with Advanced NSCLC—Real World Efficacy
Previous Article in Journal
Tunnelled Peritoneal Catheter for Malignant Ascites—An Open-Label, Prospective, Observational Trial
Previous Article in Special Issue
Mutations Associated with No Durable Clinical Benefit to Immune Checkpoint Blockade in Non-S-Cell Lung Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Classification of Non-Small Cell Lung Cancer’s Tumor Immune Micro-Environment and Strategies to Augment Its Response to Immune Checkpoint Blockade

1
Department of Radiation Oncology, Beijing Chest Hospital, Capital Medical University, Beijing 101100, China
2
Department of Radiation Oncology, Jiangsu Cancer Hospital, Nanjing Medical University, Nanjing 210009, China
3
Center for Statistical Science, Tsinghua University, Beijing 100084, China
4
Department of Radiation Oncology, Howard University, Washington, DC 20060, USA
5
Department of Radiation Oncology, China-Japan Friendship Hospital, Beijing 100029, China
6
Division of Thoracic Surgery, Department of Surgery, University of California at Los Angeles, Los Angeles, CA 90095, USA
*
Authors to whom correspondence should be addressed.
Cancers 2021, 13(12), 2924; https://doi.org/10.3390/cancers13122924
Submission received: 23 April 2021 / Revised: 3 June 2021 / Accepted: 9 June 2021 / Published: 11 June 2021
(This article belongs to the Special Issue Immunotherapy for Non-small Cell Lung Cancer)

Abstract

:

Simple Summary

Immune checkpoint blockade (ICB) has become a major treatment for lung cancer. Better understanding of the tumor immune micro-environment (TIME) in non-small cell lung cancer (NSCLC) is urgently needed to better treat it with this type of therapy. In this review, we describe and explore how NSCLC’s TIME relates to response to ICB, as well as how to treat those with unresponsive types of TIME, which will significantly impact future research in lung cancer immunotherapy.

Abstract

Immune checkpoint blockade (ICB) with checkpoint inhibitors has led to significant and durable response in a subset of patients with advanced stage EGFR and ALK wild-type non-small cell lung cancer (NSCLC). This has been consistently shown to be correlated with the unique characteristics of each patient’s tumor immune micro-environment (TIME), including the composition and distribution of the tumor immune cell infiltrate; the expression of various checkpoints by tumor and immune cells, such as PD-L1; and the presence of various cytokines and chemokines. In this review, the classification of various types of TIME that are present in NSCLC and their correlation with response to ICB in NSCLC are discussed. This is conducted with a focus on the characteristics and identifiable biomarkers of different TIME subtypes that may also be used to predict NSCLC’s clinical response to ICB. Finally, treatment strategies to augment response to ICB in NSCLC with unresponsive types of TIME are explored.

1. Introduction

The systemic therapeutic options for advanced-stage non-small cell lung cancer (NSCLC) have expanded greatly in recent years to include not only chemotherapy and targeted therapies but also immune checkpoint inhibitors (ICI) [1]. Clinical outcome in patients with PD-L1 expressing treatment-naïve stage IV or previously-treated NSCLC has significantly improved with the emergence of anti-PD-1 and anti-PD-L1 ICIs [2,3,4,5]. In the first-line setting, significant survival advantage over standard chemotherapy with anti-PD-1/anti-PD-L1(anti-PD-(L)1) monotherapy has been consistently observed in EGFR and ALK wild-type stage IV patients with tumor cell PD-L1 expression ≥ 50% [4,5]. For those with PD-L1 expression < 50%, combining an anti-PD-1 antibody with standard chemotherapy has become the first-line treatment of choice on the basis of both superior progression-free survival (PFS) and overall survival (OS) observed over standard chemotherapy in randomized controlled phase 3 trials [6,7]. In previously-treated EGFR and ALK wild-type patients with any PD-L1 expression, a survival advantage over chemotherapy from anti-PD-(L)1 monotherapy was also consistently found [8,9,10]. This advantage over chemotherapy appears to be largest in patients with high PD-L1-expressing tumors (tumor cells: ≥50%, or tumor infiltrating immune cells: ≥10%). In addition, durable response significantly longer than that of chemotherapy was observed in responders to anti-PD-(L)1 antibodies [9]. Overall, the majority of current clinical evidence demonstrated that EGFR and ALK wild-type advanced-stage NSCLC patients with high PD-L1-expressing tumors benefited the most from anti-PD-(L)1 ICIs, despite quantitative variations between the currently available PD-L1 immunohistochemistry (IHC) assays [5,11]. However, PD-L1 expression level alone does not always predict for response to anti-PD-(L)1 ICIs [12]. Independent from PD-L1, a high tumor mutational burden (TMB), which correlates with tumor neoantigen load and effector T cell interferon (IFN)-γ gene signatures, was also shown to correlate with therapeutic benefit from ICIs [5,12,13,14,15,16]. PD-L1 expression level, TMB, or effector T cell IFN-γ gene signatures may each correlate with certain characteristics of a tumor immune micro-environment (TIME) that will be optimal for PD-(L)1 immune checkpoint blockade (ICB). However, none of them alone can be used to reliably select for all responders to anti-PD-(L)1 ICIs. More thorough understanding of NSCLC’s TIME is required in order to select NSCLC patients more reliably for ICIs. In this review, the classification of different types of TIME that may exist in NSCLC and their characteristics are discussed in the context of NSCLC’s response to ICB. Furthermore, strategies to augment ICI’s therapeutic efficacy in NSCLC patients who respond poorly are explored.

2. TIME Classification Applicable to NSCLC and Its Correlation with Response to ICB

One of the major immune-inhibitory mechanisms in the tumor micro-environment is the upregulation of PD-1 expression in tumor-infiltrating lymphocytes (TILs), leading to CD8+ T cell suppression and regulatory T (Treg) cell proliferation upon interaction with its ligands (PD1 ligands 1 and 2: PD-L1 and PD-L2, respectively), which are upregulated on tumor cells through constitutive oncogenic signaling, or an adaptive response to interferon signaling-triggered antitumor immunity [17]. Because of this underlying mechanism, antitumor activity of the TILs can be restored through PD-(L)1 immune checkpoint blockade, and this has led to durable response in a subset of patients with different solid tumors [18,19]. In NSCLC’s tumor microenvironment (TME), PD-L1 can be expressed in tumor and/or immune cells. Interestingly, response to anti-PD-L1 antibody has been correlated with PD-L1 expression in tumor-infiltrating immune cells, but not in tumor cells [19]. This is likely related to the removal of myeloid cell-mediated immune suppression, leading to increased T cell activation resulting from enhanced antigen presentation upon PD-(L)1 blockade [20,21,22,23]. The TIME of poor responders to anti-PD(L)1 therapy has initially been characterized into the following types on the basis of histological observations before and after treatment with an anti-PD-L1 antibody: little or no tumor-infiltrating immune cells (immunological ignorance), intra-tumoral immune cell infiltration with minimal or no PD-L1 expression (a non-functional immune response), and an excluded immune infiltrate around the outer edge of the tumor cell cluster [19]. These types of TIME have no evidence of functional effector T cells (Table 1).
Another TIME classification system that also applies to NSCLC has been proposed [24]. In this system, the TIME is classified by the level of tumor PD-L1 expression and TILs: type I, PD-L1+ and TILs+; type II, PD-L1- and TILs-; type III, PD-L1+ and TILs-; type IV, PD-L1- and TILs+ (Table 1). Type I TIME is consistent with a state of adaptive immune resistance with T cell exhaustion mediated by the PD-1–PD-L1 inhibitory immune axis, which has been effectively targeted with anti-PD-(L)-1 blockade. Here, PD-(L)1 expression in the tumor-infiltrating CD8+ T cells has been essential to PD-(L)1 ICI therapeutic efficacy [19,25,26]. Type II TIME, which represents a state of immunological ignorance, has been associated with a lack of response to ICB [19,24]. Type III TIME represents a state of constitutive PD-L1 expression on tumor cells resulting from oncogenic signaling pathway activation, which is more prevalent in oncogenic mutation-driven cancers, such as adenocarcinoma of the lung (LUAD). Increased PD-L1 expression has been observed on NSCLC cells with activating gene alterations in KRAS, EGFR, and ALK, which has been associated with upregulated MAPK, PI3K–AKT–mTOR signaling, and JAK–STAT3 activation [27,28,29,30,31,32,33]. However, such expression is not due to the presence of functional TILs [34]. Subsequently, response to anti-PD-(L)1 ICIs alone is poor, despite PD-L1 expression in tumor cells. This has been reported in NSCLC patients with EGFR mutations and ALK rearrangements, which are also associated with low tumor neoantigen load [35,36]. Type IV TIME describes a state of ineffective IFN-γ signaling that fails to induce any PD-L1 expression [37], or an environment of immune exhaustion through additional immune checkpoints. For NSCLC, alternative immune checkpoints, such as B7x and HHLA2, were found to be expressed in the majority of PD-L1-negative cases, which inhibited T cell receptor (TCR)-mediated CD4+, CD8+ T cell proliferation, and T cell cytokine production [38].
The four-type classification system captures the main features of a TIME responsive to PD-(L)1 immune check point blockade, a state of adaptive immune resistance or T cell exhaustion that relies heavily on the PD-(L)1 immune checkpoint: increased PD-(L)1 expression on tumor and immune cells, and prominent tumor infiltration by functional TILs. This type of TIME is also described as an “inflamed” TIME. On the other hand, the main feature of an unresponsive or “cold” TIME is a lack of functional TILs in the TIME, which can be characterized with a lack of TILs (type II: immunological ignorance, excluded infiltrate, or type III: intrinsic induction), or the presence of non-functional TILs (type IV: tolerance; non-functional immune response). These types of TIMEs are associated with or without PD-L1 expression, which further demonstrates the limitations of using PD-L1 expression alone to select patients for anti-PD-(L)1 ICIs and a need for treatment strategies to augment tumor response to ICIs in cancers with an unresponsive TIME. Overall, different TIME subtypes represent variations in different aspects or steps of antitumor immunity generation and maintenance, involving a variety of factors that are intrinsic to tumor cells and extrinsically present in the TME. They will all need to be further understood in order to better characterize the TIME and effectively target tumors with unresponsive types of TIME [39,40].

3. TIME Subtype Classification Based on Analysis of Immunogenomic Data from the Cancer Genome Atlas (TCGA)

To further understand the cancer immune landscape, researchers used various immunogenomic methods to classify the TIME across 33 cancers into the wound-healing, IFN-γ-dominant, inflammatory, lymphocyte-depleted, immunologically quiet, and TGF-β-dominant subtypes on the basis of the distinct distribution of five immune-oncologic gene signatures (macrophages/monocytes, lymphocyte infiltrate, TGF-β response, IFN-γ response, and wound healing) [41]. Their characteristics are summarized in Table 2.
The wound healing, IFN-γ-dominant, and inflammatory subtypes are associated with relatively higher lymphocyte fractions (LF), which is the highest in the IFN-γ-dominant TIME. Type II helper T cells (Th2) and regulatory T cells (Tregs) were also elevated in the wound healing and IFN-γ-dominant TIME subtypes, as observed in a TGF-β-dominant TIME. The lymphocyte-depleted, immunologically quiet, and TGF-β-dominant TIME subtypes are associated with noticeably higher fractions of M2 macrophages and lower fractions of M1 macrophages. The highest and lowest M1/M2 ratios were observed in the IFN-γ-dominant and the immunologically quiet subtypes, respectively. Overall, the inflammatory subtype was associated with the best overall survival (OS). Only increased LF in the wound healing and IFN-γ dominant TIMEs significantly correlated with increased OS. This was likely related to the lower tumor proliferation rate associated with the inflammatory TIME. The lymphocyte-depleted, immunologically quiet, and TGF-β-dominant subtypes were associated with lower LF, worse survival, and higher incidence of progression. Factors associated with increased immune activation, such as lymphocyte infiltration, TCR richness, and increased fractions of Th17 and Th1 cells are associated with improved survival, while features of immune suppression, such as the wound healing (high angiogenic gene expression), macrophage regulation, and TGF-β signatures are associated with shortened survival [41].
The proportions of different TIME subtypes vary substantially among different cancers. The inflammatory, IFN-γ-dominant, and wound-healing subtypes are most common in lung adenocarcinoma (LUAD), while wound-healing and IFN-γ-dominant subtypes predominate in lung squamous cell carcinoma (LUSC). The immunologically quiet TIME is absent in both LUAD and LUSC. Consistent with their predominant TIME subtypes, LUAD and LUSC have the highest leukocyte fractions among all solid tumors analyzed, which partially explains their response to ICIs [9,41,42,43]. Increases in lymphocyte and macrophage signatures are associated with increased OS for LUAD and prolonged progression-free interval (PFI) for both LUAD and LUSC. This is most likely related to the increased fractions of CD8+ T cells and M1 macrophages in their predominant TIME subtypes. When broken down to specific immune cells, monocytes, mast cells (resting), dendritic cells (DCs), and memory B cells are prominently associated with prolonged OS for LUAD, whereas Tfh cells, γδ T cells, CD8+ T cells, activated NK cells, and M1 macrophages are associated with prolonged OS for LUSC. Tregs, CD8+ T cells, CD4 T cells, resting mast cells, M1 macrophages, DCs (resting), and memory B cells are associated with prolonged PFI for both LUAD and LUSC, thus suggesting the importance of an overall active immune infiltrate for achieving a durable response and prolonged survival after ICB in lung cancer patients.
The tumor neo-antigen load is highest in the wound healing and IFN-γ dominant TIMEs and lowest in the immunologically quiet TIME. Higher tumor neo-antigen loads in the first two types of TIMEs are associated with increased PFI, but the opposite has been observed in the inflammatory, lymphocyte-depleted, and immunologically quiet TIME subtypes [41]. This finding may relate to the presence of a normal adaptive antitumor immune response to increased tumor neo-antigens in the first two TIME subtypes but the presence of immune tolerance and immunological ignorance/exclusion in the latter three TIME subtypes. The way in which the level of tumor neoantigens associates with the level of TILs in each TIME subtype remains to be further investigated. Among all factors of immunogenicity, elevated SNV neoantigen load, non-silent mutations, and intra-tumoral heterogeneity (ITH) generally correlate with increased leukocyte fraction within the TIME. This usually represents elevated CD8+ T cells, M1 macrophages, and CD4+ memory T cells, and decreased Treg, mast, DC, and memory B cells. These correlations are strongest for in an inflammatory TIME, with weaker correlations observed in the wound healing, IFN-γ dominant, and the lymphocyte depleted TIMEs.
Different levels of driver mutation enrichment are found in different TIME subtypes, with most of them identified in the wound healing and IFN-γ dominant TIMEs, which are also predominant TIME subtypes in LUSC and LUAD. These alterations are associated with different levels of tumor neoantigens and/or the expression of various immunomodulators (IMs) (Table 3).
Some are associated with increased leukocyte fraction (TP53, HLA-B, BRAF, PTEN, NF1, APC, and CASP8), while others are associated with decreased leukocyte fraction (IDH1 R132H, GATA3, KRAS, NRAS, CTNNB1, and NOTCH1). Their association with tumor neoantigen generation, IM expression, and ultimately leukocyte fraction provides further evidence for tumor intrinsic gene alterations’ role in the sculpting of the TIME, which warrants further exploration to guide the treatment of NSCLC and other solid tumors [41].
The pattern of IM expression varies in different TIME subtypes. Stimulatory modulator CXCL10 is most highly expressed in the IFN-γ-dominant TIME, while inhibitory modulators, such as EDNRB and BTLA, are most highly expressed in the more immune-suppressive TIME subtypes. A balance between T cell activation and suppression is found in more immune-stimulatory TIME subtypes, which is evidenced by the expression of both stimulatory and inhibitory IM genes, such as SLAMF7, TNFSF4 (OX40L), IL10, CD40, and IDO1. On the contrary, modulators associated with immune infiltration are more frequently deleted in the immunologically quiet TIME (e.g., TGFB1, KIR2DL1, KIR2DL3), which is consistent with a lack of TILs in this TIME subtype. Overall, TIME subtypes with increased CD8+ T cell infiltration have been associated with the expression of stimulatory IMs, while those with increased infiltration by CD4 T cells and macrophages were associated with increased TGF-β signaling (Table 2). This pattern of IM expression reflects the predominance of different extracellular signaling networks associated with the fraction of different immune cells in the TIME [41].
Intrinsic tumor mutations interact with external signaling networks in a particular TIME with different driver mutations modulating IM expression in a TIME subtype-specific manner through common transcription factors (TFs). For example, ATM mutations and co-occurring STK11 and SMARCA4 mutations may drive wound healing TIME-specific gene expression through STAT5A in LUAD, while KEAP1 mutations, which often co-occur with STK11 and SMARCA4 mutations, drive the expression of genes specific to the immunologically quiet and TGF-β-dominant TIMEs through IRF8 in LUAD [41,44]. In LUSC, NFE2L2 mutation may drive the expression of wound healing and IFN-γ-dominant TIME-specific genes through IRF4, as well as the TGF-β dominant TIME specific gene expression through NFKB2 [41]. TIME characterization may be further enhanced with identifying T cell associated receptors and ligands that are uniquely present or absent in particular TIME subtypes, such as the absence of CTLA, LAG-3, TIM-3, TIGIT, ICOS, and IL2A in the inflammatory TIME, or the presence of IL1B and VEGFB in the TGF-β dominant TIME [41].

4. Potential Biomarkers and Therapeutic Targets in ICI Responsive vs. Nonresponsive TIME with a Focus on NSCLC

The presence of functional TILs is critical to antitumor immunity, and response to ICI largely rests on the presence of a TIME with increased TILs, as well as increased expression of PD-(L)1 in both tumor and immune cells, especially early during treatment [19,45,46,47,48]. Additional features include increased CD4 T cells, M1/M2 ratio, IFN-γ signaling, tumor neoantigen load/TMB, and upregulation of various stimulatory and inhibitory IMs [19,47,48,49,50,51,52,53]. These features are most consistent with that of an IFN-γ dominant TIME, while many of them are also present in the inflammatory and the wound healing TIMEs (Table 2). These TIME subtypes represent the predominant TIME subtypes in NSCLC. They are also enriched with many driver mutations, many of which are associated with increased leukocyte fraction within the TIME (Table 3). All these features may serve as biomarkers to identify TIME subtypes that are associated with response to ICI in NSCLC.
As a result of tumor-driven tolerance, poor antitumor immunity and a lack of response to ICI may be observed, even in the presence of tumor-associated antigen (TAA)-specific T cells [54]. This may be due to tumor-induced T cell apoptosis, T cell suppression by suppressive cytokines, such as TGFβ, or suppressive immune cells, such as Tregs or myeloid-derived suppressor cells (MDSCs), as well as altered expression of stimulatory or inhibitory immune checkpoints or modulators on T cells and other immune cells [54,55,56,57,58,59,60,61]. In NSCLC, tolerance commonly occurs with low or no PD-L1 expression on tumor cells due to impaired IFN signaling, or the activation of alternative immune checkpoints [62,63]. The impaired IFN-γ signaling can be associated with inactivating JAK mutations, which may be enriched in a wound healing TIME subtype (Table 3). These mutations result in impaired MHC I upregulation, tumor cell proliferation, and poor response to anti-PD-(L)1 blockade, partly attributing to a paucity of tumor cell PD-L1 expression [37,64]. Therefore, inactivating JAK mutations may serve as a biomarker for poor response to ICI in the presence of TILs. This may be especially useful in LUSC or LUAD, which often present with a wound-healing TIME. They are also targetable therapeutically, as JAK-independent mechanisms of pro-inflammatory pathway activation can be induced. For instance, activation of the STING pathway can be induced by STING agonists, chemotherapy, or radiotherapy in the presence of intra-tumoral dendritic cells [65,66,67,68]. Dysfunctional TILs in the absence of tumor PD-L1 expression may also represent increased sub-population of terminally exhausted CD8+ T cells, which are immune-suppressed through multiple immune checkpoints other than the PD-(L)1 checkpoint [38,69,70,71,72,73,74,75,76,77]. These terminally exhausted CD8+ cells may be identified by their expression of multiple immune checkpoints and modulators other than PD-(L)1 [73,74,75,76,77,78,79,80]. In TIME’s possession of many characteristics of immune tolerance (Table 2 and Table 3), such as the TGF-β-dominant and wound healing TIMEs, combining inhibitors to these immune checkpoints and modulators may be a viable treatment strategy, which has been shown to be feasible pre-clinically with the simultaneous blockade of PD-1 and TIM-3 [81].
A paucity of TILs with or without PD-L1 expression in the TME, a state of “immunological ignorance” or “excluded infiltrate”, has been associated with a poor response to ICIs. Such a state is most consistent with an immunologically quiet TIME, which is often associated with slowly proliferating tumors, or a lymphocyte depleted TIME, which is more often observed in fast proliferating tumors, such as NSCLC [82]. These TIME subtypes are mainly characterized by the lowest levels of any immune infiltrate with the lowest fraction of TILs but the highest fractions of macrophages (highest M2/M1 ratio). In addition, they are associated with relatively lower level of tumor neoantigen load, TCR diversity, CXCL10, and the highest level of EDNRB (Table 2), which are suggestive of an overall immunosuppressive TME with poor immunogenicity and T cell trafficking. IDH1 mutation, a mutation commonly associated with a non-inflamed immune phenotype, is most commonly found in the immunologically quiet and the lymphocyte depleted TIMEs [83]. It accounts for the majority of mutation-associated neoantigens (MANAs) in these TIME subtypes. EGFR mutation is enriched in the lymphocyte-depleted TIME, which has been associated with low baseline PD-L1 expression and low levels of functional cytotoxic T lymphocytes, despite a state of constitutive PD-L1 expression in the presence of activating EGFR mutations [27,34,84]. Other mutations associated with poor T cell infiltration, such as mutations in the Wnt/β-catenin pathway, have also been identified in these TIME subtypes [85,86,87,88]. These mutations, along with TIME composition, may help identify immunologically ignorant/excluded TIME subtypes. However, the immunologically quiet TIME largely pertains to slow-growing gliomas and was not identified in the TCGA NSCLC samples [41].
Immunological ignorance/exclusion may result from a lack of tumor neoantigens, as well as impairments in antigen presentation, along with T cell priming/activation, tumor trafficking, or infiltration [40,89]. These mechanisms of immune suppression are commonly identified in NSCLC. Despite a relatively high tumor-associated antigen (TAA) load associated with NSCLC at baseline, its TAA profile may change overtime and after treatment as a result of immune editing due to the loss of tumor subclones or chromosomal loss of truncal mutations, leading to the elimination of immunity-generating clonal TAAs [90,91,92,93]. Immune editing in NSCLC may also be associated with the presence of intrinsic tumor mutations. For instance, the lack of an adaptive immune response in EGFR mutant LUAD has been associated with low levels of TAAs and T cell clonality, leading to poor T cell priming and activation [94]. Wnt/β-catenin activation due to mutations, somatic copy number alterations (SCNAs), or over-expression has also been identified in NSCLC, leading to immunological ignorant/exclusion caused by impaired antigen presentation, T cell priming/activation, and trafficking [85,86,87,88,95,96]. As shown in melanoma, β-catenin activation within tumor cells results in transcription repressor ATF3-induced suppression of CCL4 expression, causing poor DC recruitment and cross-presentation in the lymph nodes, as well as a lack of CXCL9/CXCL10-secreting DCs at the tumor site, leading to poor CD8+ effector T cell recruitment [86,95]. Moreover, β-catenin activation in DCs inhibits T cell priming by upregulation of mTOR-dependent IL-10 production [87]. However, β-catenin in DCs is required for T cell maintenance after clonal expansion.
Other causes of an immunologically ignorant/exclusive TIME, including deficits in tumor antigen presentation/tumor cell recognition through the loss of MHC I expression and/or the downregulation or inactivation of beta-2 microglobulin (B2M), a component of the MHC class I complex in the TME; impaired T cell extravasation and tumor infiltration through various routes of chemokine modulation; and tumor intrinsic factors, such as galectin-1, are also identified in NSCLC [97,98,99,100,101,102,103,104,105,106,107]. These impairments are largely caused by gene alterations in tumor intrinsic signaling pathways, some of which lead to a TIME with constitutive tumor PD-L1 expression but impaired T cell infiltration [108]. They include methylation of IFN-β-related genes, EGFR mutations, ALK re-arrangements, mutations or other types of gene alterations in the WNT/β-catenin pathway, PTEN loss, STK11/LKB1 mutations, KEAP1/NFE2L2 mutations, Myc over-expression, and rarely IDH1 mutations [27,28,29,30,31,32,33,34,35,44,85,86,94,96,101,108,109,110,111,112,113,114,115]. On the contrary, SMARCA4, TP53, and KRAS mutations have been associated with increased TILs and improved response to anti-PD-(L)1 in LUAD [116,117,118,119,120]. Their identification can not only enhance the characterization of each patient’s TIME but also aid in developing therapeutic strategies that may augment ICI efficacy in NSCLC [121,122].
Various cytokines have been shown to have an active role in T cell exclusion and immune suppression [123]. Among them, VEGF and TGF-β are commonly known to play a role in lung cancer progression [124,125]. VEGF induces T cell apoptosis through the stimulation of FasL expression in endothelial cells; inhibits T cell recruitment and adhesion through inhibiting TNFα-mediated expression of ICAM1, VCAM1, CXCL10, CXCL11; and stimulates the expression of suppressive immune checkpoints, such as PD-L1, CTLA4, TIM-3, and LAG-3 [102,126,127,128]. In addition, it is associated with the inhibition of DC maturation and increased Tregs and MDSCs in the TIME [129]. However, VEGF-induced T cell exclusion can be reversed by antiangiogenic agents, which leads to vessel normalization and increased T cell infiltration [127,130]. Increased angiogenesis does not correlate with the magnitude of tumor infiltration by T cells. As shown in the TCGA data, elevated angiogenesis may occur in both the wound healing and the lymphocyte-depleted TIME subtypes (Table 2), thus reflecting the co-existence of multiple mechanisms of immune suppression by VEGF and other angiogenic factors. This also makes VEGF and angiogenesis an important therapeutic target to enhance ICI’s efficacy, which has already been shown to be effective in NSCLC [131]. Another cytokine that also plays a critical role in immune suppression through multiple mechanisms, the TGF-β, is associated with a state of excluded immune infiltrate in the tumor stroma upon treatment with anti-PD(L)1 ICI, attributed to increased TGF-β signaling in peritumoral stromal cells [132,133,134]. Integrin αvβ8 expression on tumor cells may also lead to poor T cell infiltration through the activation of the TGF-β signaling pathway within suppressive immune cells, such as macrophages [135]. TGB-β-induced suppression of CD8+ T cells by other immune cells may be more commonly observed in the inflammatory and TGF-β-dominant TIME subtypes, which are associated with strong TGF-β signaling in various immune cells, such as CD4+ T cells, B cells, macrophages, and neutrophils, as well as relative abundance of M2 macrophages and Tregs. Increased TGF-β expression by CD8+ T cells in an IFN-γ-dominant TIME may also occur, possibly as a mechanism to balance increased IFN signaling [41]. Overall, elevated VEGF and TGF-β expression may potentially serve as biomarkers of not only T cell exclusion but also an overall state of immune suppression with increased infiltration of suppressive immune cells (Table 2) [129,136]. In addition, they may serve as potential therapeutic targets to augment response to ICI in NSCLC and other malignancies [128,130,131,132,133,134,137].

5. Therapeutic Strategies to Augment Response to ICI in NSCLC with “Unresponsive” Types of TIMEs

Overall, response to anti-PD-(L)1 ICI is dependent on the presence of a state of adaptive immune resistance resulting primarily from the activation of the PD-(L)1 immune checkpoint. This often manifests as a “hot or inflamed” TIME with high PD-L1 expression and effector CD8+ T cell infiltration. As in many other cancers, a lack of response in NSCLC is observed when the TIME is in a state of immune tolerance, ignorance, or T cell exclusion, which manifests as a lack of functional T cells or a paucity of any T cells in the TIME with or without PD-L1 expression on tumor cells (Figure 1).
These cold TIME subtypes usually result from defects in different steps of antitumor-immunity generation resulting from tumor-intrinsic mutations, extrinsic factors such as suppressive cytokines and/or immune cells, or their interactions, depending on the specific tumor type and TIME subtype [40,44]. Identifying tumor features, such as the TIME subtype, particular mutations or gene alterations, and other biological features will most effectively predict patients’ response to ICI. Furthermore, it will guide the development of the most effective treatment strategy, which may maximize the efficacy of ICIs in cancer patients. This approach is especially helpful in NSCLC as its TIME is composed primarily of wound-healing, IFN-γ-dominant, or inflammatory subtypes. They are associated with high leukocyte infiltration, frequent genomic alterations/mutations, and other factors related to defects in antitumor-immunity generation and immune evasion [40,41,121,122]. Some of these characteristics can be used to both identify a cold tumor phenotype and be targeted therapeutically to augment the efficacy of ICIs [138,139,140,141].
In NSCLC, immune tolerance commonly results from loss-of-function JAK mutations; T cell exhaustion/apoptosis; increased expression of suppressive modulators and cytokines, such as IDO1 and TGF-β; and the suppression of normal CD8+ T cells’ interaction with tumor cells by Tregs and MDSCs [55,56,57,58,59,60,61,63,64,71,72,73,74,75,76,77,78,79,80,81]. These impairments reflect a state of T cell suppression/exhaustion and poor tumor cell recognition. They may be present in TIME subtypes with various magnitudes of CD8+ T cell infiltration or enriched with immunosuppressive cytokines, such as the IFN-γ-dominant or the TGF-β-dominant subtypes. When identified with a specific TIME subtype, treatment strategies combining ICI with other therapeutic agents, treatment modalities, or using other treatment approaches for immune activation may be developed to maximize antitumor response. For instance, activating the cGAS-STING pathway with radiotherapy, chemotherapy, STING agonists, or double-stranded (ds) DNA sensing-targeted nano-therapy along with adoptive T cell therapy may be considered in the presence of loss of function JAK mutations [64,65,66,67,68,141,142,143]. Among them, adoptive T cell therapy may also be combined with ICIs to provide a therapeutic solution to FasL-induced T cell apoptosis [144]. Cancer progression has been associated with increased terminal T cell exhaustion through additional immune checkpoints, such as LAG-3, TIM-3, TIGIT, and BTLA [73,74,75,76,77]. Inhibitors of these immune checkpoints have been shown to have antitumor activity and may improve treatment response in NSCLC with an overall unresponsive TIME if combined with anti-PD-(L)1 antibodies, other ICIs, or other treatment modalities [81,145,146,147,148,149,150,151]. Such a strategy can also be adopted when increased presence of suppressive modulators or immune cells is encountered [152,153,154,155,156,157,158,159,160]. Therefore, a combination strategy can effectively attack multiple immune defects simultaneously, which warrants further clinical investigations in NSCLC with tolerant TIME subtypes (Table 4).
NSCLC with an immunologically ignorant/exclusion TIME has mainly been associated with immune editing, driver mutations, and gene alterations intrinsic to the tumor, as well as the presence of immune suppressants in the TIME. This reflects a strong influence of tumor-intrinsic factors on the TIME, as well as interactions between tumor-intrinsic factors and extrinsic signaling networks within the TME. Therefore, combining ICI with therapeutics targeting these factors may also be effective in this setting (Table 4). Among them, a lack of neoantigens due to immune editing can be partially overcome by cytotoxic therapy, such as chemotherapy or radiotherapy [138,141,161,162]. These treatments induce increased TAA generation and IFN-β production by tumor cells and DCs through STING pathway activation, leading to increased MHC I expression, DC recruitment, maturation, and antigen cross-presentation [141,163,164,165]. Radiotherapy has also been shown to broaden the TCR repertoire, enhance T cell trafficking, and normalize tumor vasculature at low doses [162]. These properties have made combining chemotherapy and/or radiotherapy with ICI a major treatment strategy for NSCLC, which has led to significant improvement in clinical outcome over traditional standard of care in the advanced setting [6,7,166,167,168,169]. Poor T cell recruitment due to increased VEGF-related angiogenic signaling may potentially be reversed by combining ICIs with VEGF inhibitors, COX inhibitors, and/or FasL antibodies, which will reduce T cell apoptosis, enhance T cell trafficking, and induce high endothelial venule formation [102,126,127,128,129,130]. Similarly, TGF-β inhibitors may reverse T cell exclusion due to cancer-associated fibroblast (CAF)-mediated TGF-β signaling in addition to reducing suppressive immune cells and their activity [134,152,170]. This makes them good therapeutic options to combine with ICI and other treatment modalities in NSCLC or other malignancies whenever increased TGF-β signaling is detected in the TME [170,171].
Somatic mutations are frequent in NSCLC [172]. Many of them play a significant role in sculpting the TIME of NSCLC [14,34,35,44,85,86,94,96,101,108,109,110,111,112,113,114,115,173]. Among them, alterations in EGFR, PIK3CA, STK11, and KEAP1 are most commonly found in LUAD, while PTEN loss and PIK3CA driver mutations have been more commonly observed in LUSC [122]. Also identified in LUAD, ALK re-arrangement has been associated with significantly poorer response to ICI when compared with other commonly known driver mutations [174]. RET re-arrangements have also been associated with poor response to ICI, which is likely due to the low TMB and PD-L1 expression observed in RET mutant LUAD [175]. However, this does not preclude RET-re-arranged patients from responding to ICIs [176]. In LUAD, STK11/LKB1 and KEAP1 mutations frequently co-occur with KRAS mutations and with each other, while CTNNB1 and PIK3CA mutations are associated with EGFR mutations [177]. STK11/LKB1 mutations in KRAS mutant LUAD have been associated with poor response to anti-PD-(L)1 ICI and other cytotoxic treatments, which may be associated with STK11/LKB1 and KEAP1 co-mutations’ association with STING suppression and metabolic reprogramming with enhanced glutamine dependence in KRAS mutant LUAD [44,111,112,178,179,180,181,182,183]. Glutaminase inhibition and KEAP1 expression have been shown to enhance radiosensitivity of KRAS mutant LUAD with STK11/LKB1 and KEAP1 mutations [184,185]. Whether these potential therapeutic options can potentiate sensitivity to ICI and the synergistic effect of combing ICI with cytotoxic therapies such as radiotherapy in KRAS mutant LUAD with STK11/LKB1 and/or KEAP1/NRF2 mutations warrants further exploration [166,184,185]. Other treatment options, such as an NRF2 inhibitor, can also be added into the treatment regimen for this very aggressive subgroup of LUAD that responds poorly to any treatments [186]. A similar approach targeting the Wnt/β catenin or the PI3K–AKT–mTOR pathways along with the PD-(L)1 pathway may be taken in the presence of CTNNB1 or PIK3CA mutations [29,187,188]. However, the majority of PIK3CA mutations are associated with other mutations [189]. Among them, EGFR mutations are known to be associated with poor immunogenicity, T cell tumor infiltration, and a lack of response to ICIs [34,94,174,190,191,192]. Similarly, ALK-re-arranged LUAD responds poorly to ICI, with only increased Treg in the TIME upon the development of TKI resistance or the treatment of TKI followed by ICI [35,174,193].
Different EGFR mutations may influence the TIME in LUAD differently, leading to variations in clinical response to ICI. For instance, EGFR exon 19 deletions have been associated with lower TMB than exon 21 L858R point mutation, and this has correlated with poorer response to ICI [192]. Moreover, T790M positivity has been associated with less PD-L1 expression and shorter PFS after ICI monotherapy in LUAD [174,194]. Exon 21 mutation appears to correlate with better response to ICI, thus making combining ICI and an EGFR TKI a potential treatment strategy in EGFR mutant LUAD [195]. However, ICI administration preceding or concurrent with osimertinib has been associated with significant pulmonary toxicity [196,197]. Similarly, the clinical feasibility of combining an ALK inhibitor with an ICI is largely limited by severe hepatic toxicities [198]. On the contrary, combining ICI, cytotoxic treatment modalities, and inhibitors of other targets in the TIME, such as VEGF, may be clinically feasible and effective in NSCLC [199]. This may be partially attributed to the increased generation of TAAs with cytotoxic agents and treatment modalities, such as chemotherapy and radiotherapy [162,169]. For mutations associated with low TMB, such as alterations in EGFR, ALK, and RET, this treatment strategy could be further explored in future clinical trials.

6. Conclusions

Different TIME subtypes exist in NSCLC. The presence of functional TILs is the most critical feature of a TIME associated with antitumor response to immune checkpoint blockade. Poor response to immune checkpoint blockade is associated with a TIME with identifiable features of immune tolerance, immunological ignorance, or T cell exclusion, which can all be identified in NSCLC. These features represent defects in the generation of antitumor immunity resulting from tumor intrinsic gene alterations, TIME-specific external signaling networks, or an interaction of the two. They may be used to not only identify NSCLC with unresponsive TIME subtypes, but also to guide the development of the more effective treatments that may augment ICI’s efficacy in NSCLC with such unresponsive phenotypes. Given cytotoxic treatment, such as radiotherapy’s immunostimulatory properties that may help overcome immunological tolerance and ignorance, the way in which to best combine it with ICI should be further explored.

Author Contributions

Conceptualization: A.C., X.H., L.H., N.P.N., G.Z. Methodology: A.C. Writing—original draft preparation: A.C. Writing—review and editing: A.C., X.H., L.H., N.P.N., G.Z., R.B.C., J.M.L. Visualization: A.C. Funding acquisition: A.C., X.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

Figure 1 was created with www.biorender.com. Last accessed on 21 April 2021. L.H. acknowledges research support from China’s National Key R&D Program (grant no. 2020YFA0712400) and Shanghai Municipal Science and Technology Major Project (grant no. 2017SHZDZX01). J.M.L. acknowledges being on the Advisory Board of AstraZeneca, Bristol Myers Squibb, Genentech, and Novartis, and in the Executive/Steering Committee of Genentech and Novartis.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. National Comprehensive Cancer Network. Non-Small Cell Lung Cancer (Version 4. 2021). Available online: https://www.nccn.org/professionals/physician_gls/pdf/nscl.pdf (accessed on 10 April 2021).
  2. Herbst, R.S.; Baas, P.; Kim, D.; Felip, E.; Pérez-Gracia, J.L.; Han, J.; Molina, J.; Kim, J.; Arvis, C.D.; Ahn, M.; et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): A randomized controlled trial. Lancet 2015, 387, 1540–1550. [Google Scholar] [CrossRef]
  3. Reck, M.; Rodríguez-Abreu, D.; Robinson, A.G.; Hui, R.; Csőszi, T.; Fülöp, A.; Gottfried, M.; Peled, N.; Tafreshi, A.; Cuffe, S.; et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N. Engl. J. Med. 2016, 375, 1823–1833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Mok, T.S.K.; Wu, Y.; Kudaba, I.; Kowalski, D.M.; Cho, B.C.; Turna, H.Z.; Castro, G., Jr.; Srimuninnimit, V.; Laktionov, K.K.; Bondarenko, I.; et al. Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): A randomized, open-label, controlled, phase 3 trial. Lancet 2019, 393, 1819–1830. [Google Scholar] [CrossRef]
  5. Herbst, R.S.; Giaccone, G.; de Marinis, F.; Reinmuth, N.; Vergnenegre, A.; Barrios, C.H.; Morise, M.; Felip, E.; Andric, Z.; Geater, S.; et al. Atezolizumab for first-line treatment of PD-L1-selected patients with NSCLC. N. Engl. J. Med. 2020, 383, 1328–1339. [Google Scholar] [CrossRef]
  6. Gandhi, L.; Rodríguez-Abreu, D.; Gadgeel, S.; Esteban, E.; Felip, E.; De Angelis, F.; Domine, M.; Clingan, P.; Hochmair, M.J.; Powell, S.F.; et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N. Engl. J. Med. 2018, 378, 2078–2092. [Google Scholar] [CrossRef]
  7. Paz-Ares, L.; Luft, A.; Vicente, D.; Tafreshi, A.; Gümüş, M.; Mazières, J.; Hermes, B.; Çay Şenler, F.; Csőszi, T.; Fülöp, A.; et al. Pembrolizumab plus chemotherapy for squamous non-small-cell lung cancer. N. Engl. J. Med. 2018, 379, 2040–2051. [Google Scholar] [CrossRef]
  8. Horn, L.; Spigel, D.R.; Vokes, E.E.; Holgado, E.; Ready, N.; Steins, M.; Poddubskaya, E.; Borghaei, H.; Felip, E.; Paz-Ares, L.; et al. Nivolumab versus docetaxel in previously treated patients with advanced non-small-cell lung cancer: Two-year outcomes from two randomized, open-label, phase III trials (CheckMate 017 and CheckMate 057). J. Clin. Oncol. 2017, 35, 3924–3933. [Google Scholar] [CrossRef]
  9. Herbst, R.S.; Garon, E.B.; Kim, D.; Cho, B.C.; Perez-Gracia, J.L.; Han, J.; Arvis, C.D.; Majem, M.; Forster, M.D.; Monnet, I.; et al. Long-term outcomes and retreatment among patients with previously treated, programmed death-ligand 1-positive, advanced non-small-cell lung cancer in the Keynote-010 study. J. Clin. Oncol. 2020, 38, 1580–1591. [Google Scholar] [CrossRef]
  10. Rittmeyer, A.; Barlesi, F.; Waterkamp, D.; Park, K.; Ciardiello, F.; von Pawel, J.; Gadgeel, S.M.; Hida, T.; Kowalski, D.M.; Dols, M.C.; et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): A phase 3, open-label, multicentre randomised controlled trial. Lancet 2017, 389, 255–265. [Google Scholar] [CrossRef]
  11. Hirsch, F.R.; McElhinny, A.; Stanforth, D.; Ranger-Moore, J.; Jansson, M.; Kulangara, K.; Richardson, W.; Towne, P.; Hanks, D.; Vennapusa, B.; et al. PD-L1 immunohistochemistry assays for lung cancer: Results from phase 1 of the Blueprint PD-L1 IHC assay comparison project. J. Thor. Oncol. 2016, 12, 208–222. [Google Scholar] [CrossRef] [Green Version]
  12. Carbone, D.P.; Reck, M.; Paz-Ares, L.; Creelan, B.; Horn, L.; Steins, M.; Felip, E.; van den Heuvel, M.M.; Ciuleanu, T.-E.; Badin, F.; et al. First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N. Engl. J. Med. 2017, 376, 2415–2426. [Google Scholar] [CrossRef]
  13. Gandara, D.R.; Paul, S.M.; Kowanetz, M.; Schleifman, E.; Zou, W.; Li, Y.; Rittmeyer, A.; Fehrenhacher, L.; Otto, G.; Malboeuf, C.; et al. Blood-based tumor mutational burden as a predictor of clinical benefit in non-small-cell lung cancer patients treated with atezolizumab. Nat. Med. 2018, 24, 1441–1448. [Google Scholar] [CrossRef]
  14. 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-generatoin sequencing. J. Clin. Oncol. 2018, 36, 633–641. [Google Scholar] [CrossRef] [PubMed]
  15. Hellmann, M.D.; Ciuleanu, T.-E.; Pluzanski, A.; Lee, J.S.; Otterson, G.A.; Audigier-Valette, C.; Minenza, E.; Linardou, H.; Burgers, S.; Salman, P.; et al. Nivolumab plus ipilumumab in lung cancer with a high tumor mutational burden. N. Engl. J. Med. 2018, 378, 2093–2104. [Google Scholar] [CrossRef]
  16. 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 randomized controlled trial. Lancet 2016, 387, 1837–1846. [Google Scholar] [CrossRef]
  17. Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Ribas, A.; Wolchok, J.D. Cancer immunotherapy using checkpoint blockade. Science 2018, 359, 1350–1355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Herbst, R.S.; Soria, J.; 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] [PubMed] [Green Version]
  20. Boussiotis, V.A. Molecular and biochemical aspects of the PD-1 checkpoint pathway. N. Engl. J. Med. 2016, 375, 1767–1778. [Google Scholar] [CrossRef] [Green Version]
  21. Nakamura, K.; Smyth, M.J. Myeloid immunosuppression and immune checkpoints in the tumor microenvironment. Cell. Mol. Immunol. 2020, 17, 1–12. [Google Scholar] [CrossRef] [PubMed]
  22. Kleinovink, J.W.; Marijt, K.A.; Schoonderwoerd, M.J.A.; van Hall, T.; Ossendorp, F.; Fransen, M.F. PD-L1 expression on malignant cells is no prerequisite for checkpoint therapy. Oncoimmunology 2017, 6, e1294299. [Google Scholar] [CrossRef] [PubMed]
  23. Tang, H.; Liang, Y.; Anders, R.A.; Taube, J.M.; Qiu, X.; Mulgaonkar, A.; Liu, X.; Harrington, S.M.; Guo, J.; Xin, Y.; et al. PD-L1 on host cells is essential for PD-L1 blockade-mediated tumor regression. J. Clin. Investig. 2018, 128, 580–588. [Google Scholar] [CrossRef] [Green Version]
  24. Teng, M.W.L.; Ngiow, S.F.; Ribas, A.; Smyth, M.J. Classifying cancers based on T-cell infiltration and PD-L1. Cancer Res. 2015, 75, 2139–2145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Tumeh, P.C.; Harview, C.L.; Yearley, J.H.; Shintaku, I.P.; Taylor, E.J.M.; Robert, L.; Chmielowski, B.; Spasic, M.; Henry, G.; Ciobanu, V.; et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014, 515, 568–571. [Google Scholar] [CrossRef] [PubMed]
  26. Thommen, D.S.; Koelzer, V.H.; Herzig, P.; Roller, A.; Trefny, M.; Dimeloe, S.; Kiialainen, A.; Hanhart, J.; Schill, C.; Hess, C.; et al. A transcriptionally and functionally distinct PD-1+ CD8+ T cell pool with predictive potential in non-small-cell lung cancer treated with PD-1 blockade. Nat. Med. 2018, 24, 994–1004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Chen, N.; Fang, W.; Zhan, J.; Hong, S.; Tang, Y.; Kang, S.; Zhang, Y.; He, X.; Zhou, T.; Qin, T.; et al. Upregulation of PD-L1 by EGFR activation mediates the immune escape in EGFR-driven NSCLC: Implication for optional anti-PD-1/PD-L1 immune therapy for NSCLC patients with EGFR mutations. J. Thorac. Oncol. 2015, 10, 910–923. [Google Scholar] [CrossRef] [Green Version]
  28. Ota, K.; Azuma, K.; Kawahara, A.; Hattori, S.; Iwama, E.; Tanizaki, J.; Harada, T.; Matsumoto, K.; Takayama, K.; Takamori, S.; et al. Induction of PD-L1 expression by the EML4-ALK oncoprotein and downstream signaling pathways in non-small cell lung cancer. Clin. Cancer Res. 2015, 21, 4014–4021. [Google Scholar] [CrossRef] [Green Version]
  29. Lastwika, K.J.; Wilson, W., 3rd; Li, Q.K.; Norris, J.; Xu, H.; Ghazarian, S.R.; Kitagawa, H.; Kawabata, S.; Taube, J.M.; Yao, S.; et al. Control of PD-L1 expression by oncogenic activation of the AKT-mTOR pathway in non-small cell lung cancer. Cancer Res. 2016, 76, 227–238. [Google Scholar] [CrossRef] [Green Version]
  30. Karachaliou, N.; Rosell, R. Systemic treatment in EGFR-ALK NSCLC patients: Second line therapy and beyond. Cancer Biol. Med. 2014, 11, 173–181. [Google Scholar] [CrossRef] [Green Version]
  31. Gao, S.P.; Mark, K.G.; Leslie, K.; Pao, W.; Motoi, N.; Gerald, W.L.; Travis, W.D.; Bornmann, W.; Veach, D.; Clarkson, B.; et al. Mutations in the EGFR kinase domain mediates STAT3 activation via IL-6 production in human lung adenocarcinomas. J. Clin. Investig. 2007, 117, 3846–3856. [Google Scholar] [CrossRef] [Green Version]
  32. Song, T.L.; Nairismägi, M.; Laurensia, Y.; Lim, J.; Tan, J.; Li, Z.; Pang, W.; Kizhakeyil, A.; Wijaya, G.; Huang, D.; et al. Oncogenic activation of the STAT3 pathway drives PD-L1 expression in natural killer/T-cell lymphoma. Blood 2018, 132, 1146–1158. [Google Scholar] [CrossRef] [Green Version]
  33. Chen, N.; Fang, W.; Lin, Z.; Peng, P.; Wang, J.; Zhan, J.; Hong, S.; Huang, J.; Liu, L.; Sheng, J.; et al. KRAS mutation-induced upregulation of PD-L1 mediates immune escape in human lung adenocarcinoma. Cancer Immunol. Immunother. 2017, 66, 1175–1187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Toki, M.I.; Mani, N.; Smithy, J.W.; Liu, Y.; Phil, M.; Altan, M.; Wasserman, B.; Tuktamyshov, R.; Schalper, K.; Syrigos, K.N.; et al. Immune marker profiling and programmed death ligand 1 expression across NSCLC mutations. J. Thorac. Oncol. 2018, 13, 1884–1896. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. 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]
  36. Heigener, D.F.; Reck, M. PD-1 axis inhibition in EGFR positives: A blunt sword? J. Thorac. Oncol. 2017, 12, 171–172. [Google Scholar] [CrossRef] [Green Version]
  37. 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 Disc. 2016, 7, 188–201. [Google Scholar] [CrossRef] [Green Version]
  38. Cheng, H.; Borczuk, A.; Janakiram, M.; Ren, X.; Lin, J.; Assal, A.; Halmos, B.; Perez-Soler, R.; Zang, X. Wide expression and significance of alternative immune checkpoint molecules, B7x and HHLA2, in PD-L1-negative human lung cancers. Clin. Cancer Res. 2018, 24, 1954–1964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. 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] [Green Version]
  40. Kim, J.M.; Chen, D.S. Immune escape to PD-L1/PD-1 blockade: Seven steps to success (or failure). Ann. Oncol. 2016, 27, 1492–1504. [Google Scholar] [CrossRef] [PubMed]
  41. Thorsson, V.; Gibbs, D.L.; Brown, S.D.; Wolf, D.; Bortone, D.S.; Ou Yang, T.; Porta-Pardo, E.; Gao, G.F.; Plaisier, C.L.; Eddy, J.A.; et al. The immune landscape of cancer. Immunity 2018, 48, 812–830. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Arina, A.; Karrison, T.; Galka, E.; Schreiber, K.; Weichselbaum, R.R.; Schreiber, H. Transfer of allogeneic CD4+ T cells rescues CD8+ T cells in anti-PD-L1-resistant tumors leading to tumor eradication. Cancer Immunol. Res. 2017, 5, 127–136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Siddiqui, I.; Schaeuble, K.; Chennupati, V.; Marraco, S.A.F.; Calderon-Copete, S.; Ferreira, D.P.; Carmona, S.J.; Scarpellino, L.; Gfeller, D.; Pradervand, S.; et al. Intratumoral Tcf1+ PD-1+CD8+ T cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy. Immunity 2019, 50, 195–211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Marinelli, D.; Mazzotta, M.; Scalera, S.; Terrenato, I.; Sperati, F.; D’Ambrosio, L.; Pallocca, M.; Corleone, G.; Krasniqi, E.; Pizzuti, L.; et al. KEAP1-driven co-mutations in lung adenocarcinoma unresponsive to immunotherapy despite high tumor mutational burden. Ann. Oncol. 2020, 31, 1746–1754. [Google Scholar] [CrossRef] [PubMed]
  45. Vilain, R.E.; Menzies, A.M.; Wilmott, J.S.; Kakavand, H.; Madore, J.; Guminski, A.; Liniker, E.; Kong, B.Y.; Cooper, A.J.; Howle, J.R.; et al. Dynamic changes in PD-L1 expression and immune infiltrates early during treatment predict response to PD-1 blockade in melanoma. Clin. Cancer. Res. 2017, 23, 5024–5033. [Google Scholar] [CrossRef] [Green Version]
  46. Chen, P.; 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]
  47. Niemeijer, A.N.; Sahba, S.; Smit, E.F.; Lissenberg-Witte, B.I.; de Langen, A.J.; Thunnissen, E. Association of tumour and stroma PD-1, PD-L1, CD3, CD4, CD8 expression with DCB and OS to nivolumab treatment in NSCLC patients pre-treated with chemotherapy. Br. J. Cancer 2020, 123, 392–402. [Google Scholar] [CrossRef]
  48. Liu, Y.; Zugazagoitia, J.; Ahmed, F.S.; Henick, B.S.; Gettinger, S.N.; Herbst, R.S.; Schalper, K.A.; Rimm, D.L. Immune cell PD-L1 colocalizes with macrophages and is associated with outcome in PD-1 pathway blockade therapy. Clin. Cancer Res. 2020, 26, 970–977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Etzerodt, A.; Tsalkitzi, K.; Maniecki, M.; Damsky, W.; Delfini, M.; Baudoin, E.; Moulin, M.; Bosenberg, M.; Graversen, J.H.; Auphan-Anezin, N.; et al. Specific targeting of CD163+ TAMs mobilizes inflammatory monocytes and promotes T cell-mediated tumor regression. J. Exp. Med. 2019, 216, 2394–2411. [Google Scholar] [CrossRef] [PubMed]
  50. Ayers, M.; Lunceford, J.; Nebozhyn, M.; Murphy, E.; Loboda, A.; Kaufman, D.R.; Albright, A.; Cheng, J.D.; Kang, S.P.; Shankaran, V.; et al. IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Investig. 2017, 127, 2930–2940. [Google Scholar] [CrossRef] [PubMed]
  51. Karachaliou, N.; Gonzalez-Cao, M.; Crespo, G.; Drozdowskyj, A.; Aldeguer, E.; Gimenez-Capitan, A.; Teixido, C.; Molina-Vila, M.A.; Viteri, S.; De Los Llanos Gil, M.; et al. Interferon gamma, an important marker of response to immune checkpoint blockade in non-small cell lung cancer and melanoma patients. Ther. Adv. Med. Oncol. 2018, 10, 1–23. [Google Scholar] [CrossRef] [PubMed]
  52. Hurkmans, D.P.; Kuipers, M.E.; Smit, J.; van Marion, R.; Mathijssen, R.H.J.; Postmus, P.E.; Hiemstra, P.S.; Aerts, J.G.J.V.; von der Thüsen, J.H.; van der Burg, S.H.; et al. Tumor mutational load, CD8+ T cells, expression of PD-L1 and HLA class I to guide immunotherapy decisions in NSCLC patients. Cancer Immunol. Immunother. 2020, 69, 771–777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. 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. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015, 348, 124–128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Mapara, M.Y.; Sykes, M. Tolerance and cancer: Mechanisms of tumor evasion and strategies for breaking tolerance. J. Clin. Oncol. 2004, 22, 1136–1151. [Google Scholar] [CrossRef] [PubMed]
  55. Zhu, J.; de Tenbossche, C.G.; Cané, S.; Colau, D.; van Baren, N.; Lurquin, C.; Schmitt-Verhulst, A.; Liljeström, P.; Uyttenhove, C.; van den Eynde, B.J. Resistance to cancer immunotherapy mediated by apoptosis of tumor-infiltrating lymphocytes. Nat. Comm. 2017, 8, 1404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Horton, B.L.; Williams, J.B.; Cabanov, A.; Spranger, S.; Gajewski, T.F. Intratumoral CD8+ T-cell apoptosis is a major component of T-cell dysfunction and impedes antitumor immunity. Cancer Immunol. Res. 2017, 6, 14–24. [Google Scholar] [CrossRef] [Green Version]
  57. Batlle, E.; Massagué, J. Transforming growth factor-β signaling in immunity and cancer. Immunity 2019, 50, 924–940. [Google Scholar] [CrossRef]
  58. Paluskievicz, C.M.; Cao, X.; Abdi, R.; Zheng, P.; Liu, Y.; Bromberg, J.S. T regulatory cells and priming the suppressive tumor microenvironment. Front. Immunol. 2019, 10, 2453. [Google Scholar] [CrossRef]
  59. Ostrand-Rosenberg, S.; Fenselau, C. Myeloid-derived suppressor cells: Immune suppressive cells that impair antitumor immunity and are sculpted by their environment. J. Immunol. 2018, 200, 422–431. [Google Scholar] [CrossRef] [Green Version]
  60. Bauer, C.A.; Kim, E.Y.; Marangoni, F.; Carrizosa, E.; Claudio, N.M.; Mempel, T.R. Dynamic Treg interactions with intratumoral APCs promote local CTL dysfunction. J. Clin. Investig. 2014, 124, 2425–2440. [Google Scholar] [CrossRef]
  61. Nagaraj, S.; Gupta, K.; Pisarev, V.; Kinarsky, L.; Sherman, S.; Kang, L.; Herber, D.L.; Schneck, J.; Gabrilovich, D.I. Altered recognition of antigen is a mechanism of CD8+ T cell tolerance in cancer. Nat. Med. 2007, 13, 828–835. [Google Scholar] [CrossRef] [Green Version]
  62. Mandai, M.; Hamanishi, J.; Abiko, K.; Matsumura, N.; Baba, T.; Konishi, I. Dual faces of IFNγ in cancer progression: A role of PD-L1 induction in the determination of pro- and antitumor immunity. Clin. Cancer Res. 2016, 22, 2329–2334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Theelen, W.S.M.E.; Kuilman, T.; Schulze, K.; Zou, W.; Krijgsman, O.; Peters, D.D.G.C.; Cornelissen, S.; Monkhorst, K.; Sarma, P.; Sumiyoshi, T.; et al. Absence of PD-L1 expression on tumor cells in the context of an activated immune infiltrate may indicate impaired IFNγ signaling in non-small cell lung cancer. PLoS ONE 2019, 14, e0216864. [Google Scholar] [CrossRef] [PubMed]
  64. Marabelle, A.; Aspeslagh, S.; Postel-Vinay, S.; Soria, J. JAK mutations as escape mechanisms to anti-PD-1 therapy. Cancer Discov. 2017, 7, 128–130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Corrales, L.; Glickman, L.H.; McWhirter, S.M.; Kanne, D.B.; Sivick, K.E.; Katibah, G.E.; Woo, S.; Lemmens, E.; Banda, T.; Leong, J.J.; et al. Direct activation of STING in the tumor microenvironment leads to potent and systemic tumor regression and immunity. Cell Rep. 2015, 11, 1018–1030. [Google Scholar] [CrossRef] [Green Version]
  66. Della Corte, C.M.; Sen, T.; Gay, C.M.; Ramkumar, K.; Diao, L.; Cardnell, R.J.; Rodriguez, B.L.; Stewart, C.A.; Papadimitrakopoulou, V.A.; Gibson, L.; et al. STING pathway expression identifies NSCLC with an immune-responsive phenotype. J. Thorac. Oncol. 2020, 15, 777–791. [Google Scholar] [CrossRef]
  67. Deng, L.; Liang, H.; Xu, M.; Yang, X.; Burnette, B.; Arina, A.; Li, X.; Mauceri, H.; Beckett, M.; Darga, T.; et al. STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors. Immunity 2014, 41, 843–852. [Google Scholar] [CrossRef] [Green Version]
  68. Vanpouille-Box, C.; Alard, A.; Aryankalayil, M.J.; Sarfraz, Y.; Diamond, J.M.; Schneider, R.J.; Inghirami, G.; Coleman, C.N.; Formenti, S.C.; Demaria, S. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat. Commun. 2017, 8, 15618. [Google Scholar] [CrossRef]
  69. Miller, B.C.; Sen, D.R.; Abosy, R.A.; Bi, K.; Virkud, Y.V.; LaFleur, M.W.; Yates, K.B.; Lako, A.; Felt, K.; Naik, G.S.; et al. Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat. Immunol. 2019, 20, 326–336. [Google Scholar] [CrossRef]
  70. Kurtulus, S.; Madi, A.; Escobar, G.; Klapholz, M.; Nyman, J.; Christian, E.; Pawlak, M.; Dionne, D.; Xia, J.; Rozenblatt-Rosen, O.; et al. Checkpoint blockade immunotherapy induces dynamic changes in PD-1CD8+ tumor-infiltrating T cells. Immunity 2019, 50, 181–194. [Google Scholar] [CrossRef] [Green Version]
  71. Nirschl, C.J.; Drake, C.G. Molecular pathways: Coexpression of immune checkpoint molecules: Signaling pathways and implications for cancer immunotherapy. Clin. Cancer Res. 2013, 19, 4917–4924. [Google Scholar] [CrossRef] [Green Version]
  72. Ren, S.; Tian, Q.; Amar, N.; Yu, A.; Rivard, C.J.; Caldwell, C.; Ng, T.L.; Tu, M.; Liu, Y.; Gao, D.; et al. The immune checkpoint HVEM may contribute to immune escape in non-small cell lung cancer lacking PD-L1 expression. Lung Cancer 2018, 125, 115–120. [Google Scholar] [CrossRef]
  73. Thommen, D.S.; Schreiner, J.; Müller, P.; Herzig, P.; Roller, A.; Belousov, A.; Umana, P.; Pisa, P.; Klein, C.; Bacac, M.; et al. Progression of lung cancer is associated with increased dysfunction of T cells defined by coexpression of multiple inhibitory receptors. Cancer Immunol. Res. 2015, 3, 1344–1355. [Google Scholar] [CrossRef] [Green Version]
  74. Koyama, S.; Akbay, E.A.; Li, Y.Y.; Herter-Sprie, G.S.; Buczkowski, K.A.; Richards, W.G.; Gandhi, L.; Redig, A.J.; Rodig, S.J.; Asahina, H.; et al. Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat. Comm. 2016, 7, 10501. [Google Scholar] [CrossRef] [PubMed]
  75. He, Y.; Yu, H.; Rozeboom, L.; Rivard, C.J.; Ellison, K.; Dziadziuszko, R.; Suda, K.; Ren, S.; Wu, C.; Hou, L.; et al. LAG-3 protein expression in non-small cell lung cancer and its relationship with PD-1/PD-L1 and tumor-infiltrating lymphocytes. J. Thorac. Oncol. 2017, 12, 814–823. [Google Scholar] [CrossRef] [Green Version]
  76. Datar, I.J.; Sanmamed, M.F.; Wang, J.; Henick, B.S.; Choi, J.; Badri, T.; Dong, W.; Mani, N.; Toki, M.; Mejías, L.D.; et al. Expression analysis and significance of PD-1, LAG-3 and TIM-3 in human non-small cell lung cancer using spatially-resolved and multiparametric single-cell analysis. Clin. Cancer Res. 2019, 25, 4663–4673. [Google Scholar] [CrossRef] [PubMed]
  77. Johnston, R.J.; Comps-Agrar, L.; Hackney, J.; Yu, X.; Huseni, M.; Yang, Y.; Park, S.; Javinal, V.; Chiu, H.; Irving, B.; et al. The immunoreceptor TIGIT regulates antitumor and antiviral CD8+ T cell effector function. Cancer Cell 2014, 26, 923–937. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Schalper, K.A.; Carvajal-Hausdorf, D.; McLaughlin, J.; Altan, M.; Velcheti, V.; Gaule, P.; Sanmamed, M.F.; Chen, L.; Herbst, R.S.; Rimm, D.L. Differential expression and significance of PD-L1, IDO-1, and B7-H4 in human lung cancer. Clin. Cancer Res. 2016, 23, 370–378. [Google Scholar] [CrossRef] [Green Version]
  79. Smith, C.; Chang, M.Y.; Parker, K.H.; Beury, D.W.; DuHadaway, J.B.; Flick, H.E.; Boulden, J.; Sutanto-Ward, E.; Soler, A.P.; Laury-Kleintop, L.D.; et al. IDO is a nodal pathogenic driver of lung cancer and metastasis development. Cancer Disc. 2012, 2, 722–735. [Google Scholar] [CrossRef] [Green Version]
  80. Moon, Y.W.; Hajjar, J.; Hwu, P.; Naing, A. Targeting the indoleamine 2,3-dioxygenase pathway in cancer. J. Immunother. Cancer 2015, 3, 51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Sun, F.; Guo, Z.S.; Gregory, A.D.; Shapiro, S.D.; Xiao, G.; Qu, Z. Dual but not single PD-1 or TIM-3 blockade enhances oncolytic virotherapy in refractory lung cancer. J. Immunother. Cancer 2020, 8, e000294. [Google Scholar] [CrossRef] [PubMed]
  82. Kargl, J.; Zhu, X.; Zhang, H.; Yang, G.H.Y.; Friesen, T.J.; Shipley, M.; Maeda, D.Y.; Zebala, J.A.; McKay-Fleisch, J.; Meredith, G.; et al. Neutrophil content predicts lymphocyte depletion and anti-PD1 treatment failure in NSCLC. J. Cancer Investig. Insight 2019, 4, e130850. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Kohanbash, G.; Carrera, D.A.; Shrivastav, S.; Ahn, B.J.; Jahan, N.; Mazor, T.; Chheda, Z.S.; Downey, K.M.; Watchmaker, P.B.; Beppler, C.; et al. Isocitrate dehydrogenase mutations suppress STAT1 and CD8+ T cell accumulation in gliomas. J. Clin. Investig. 2017, 127, 1425–1437. [Google Scholar] [CrossRef]
  84. Akbay, E.A.; Koyama, S.; Carretero, J.; Altabef, A.; Tchaicha, J.H.; Christensen, C.L.; Mikse, O.R.; Cherniack, A.D.; Beauchamp, E.M.; Pugh, T.J.; et al. Activation of the PD-1 pathway contributes to immune escape in EGFR-driven lung tumors. Cancer Discov. 2013, 3, 1355–1363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Luke, J.L.; Bao, R.; Sweis, R.F.; Spranger, S.; Gajewski, T.F. WNT/β-catenin pathway activation correlates with immune exclusion across human cancers. Clin. Cancer Res. 2019, 25, 3074–3083. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Spranger, S.; Bao, R.; Gajewski, T.F. Melanoma-intrinsic β-catenin signaling prevents anti-tumor immunity. Nature 2015, 523, 231–235. [Google Scholar] [CrossRef]
  87. Fu, C.; Liang, X.; Cui, W.; Ober-Blöbaum, J.L.; Vazzana, J.; Shrikant, P.A.; Lee, K.P.; Clausen, B.E.; Mellman, I.; Jiang, A. β-Catenin in dendritic cells exerts opposite functions in cross-priming and maintenance of CD8+ T cells through regulation of IL-10. Proc. Natl. Acad. Sci. USA 2015, 112, 2823–2828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Ganesh, S.; Shui, X.; Craig, K.P.; Park, J.; Wang, W.; Brown, B.D.; Abrams, M.T. RNAi-mediated β-catenin inhibition promotes T cell infiltration and antitumor activity in combination with immune checkpoint blockade. Mol. Ther. 2018, 26, 2567–2579. [Google Scholar] [CrossRef]
  89. Bonaventura, P.; Shekarian, T.; Alcazer, V.; Valladeau-Guilemond, J.; Valsesia-Wittmann, S.; Amigorena, S.; Caux, C.; Depil, S. Cold tumors: A therapeutic challenge for immunotherapy. Front. Immunol. 2019, 10, 168. [Google Scholar] [CrossRef] [Green Version]
  90. Lee, C.; Yelensky, R.; Jooss, K.; Chan, T.A. Update on tumor neoantigens and their utility: Why it is good to be different. Trends Immunol. 2018, 39, 536–548. [Google Scholar] [CrossRef]
  91. Rosenthal, R.; Cadieux, E.L.; Salgado, R.; Bakir, A.M.; Moore, D.A.; Hiley, C.T.; Lund, T.; Tanić, M.; Reading, J.L.; Joshi, K.; et al. Neoantigen-directed immune escape in lung cancer evolution. Nature 2019, 567, 479–485. [Google Scholar] [CrossRef]
  92. Anagnostou, V.; Smith, K.N.; Forde, P.M.; Niknafs, N.; Bhattacharya, R.; White, J.; Theresa, Z.; 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]
  93. McGranahan, N.; Furness, A.J.S.; 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]
  94. Reuben, A.; Zhang, J.; Chiou, S.; Gittelman, R.M.; Li, J.; Lee, W.; Fujimoto, J.; Behrens, C.; Liu, X.; Wang, F.; et al. Comprehensive T cell repertoire characterization of non-small cell lung cancer. Nat. Comm. 2020, 11, 603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. 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. [Google Scholar] [CrossRef] [Green Version]
  96. Muto, S.; Ozaki, Y.; Yamaguchi, H.; Mine, H.; Takagi, H.; Watanabe, M.; Inoue, T.; Yamaura, T.; Fukuhara, M.; Okabe, N.; et al. Tumor β-catenin expression is associated with immune evasion in non-small cell lung cancer with high tumor mutation burden. Oncol. Lett. 2021, 21, 203. [Google Scholar] [CrossRef] [PubMed]
  97. Pereira, C.; Gimenez-Xavier, P.; Pros, E.; Pajares, M.J.; Moro, M.; Gomez, A.; Navarro, A.; Condom, E.; Moran, S.; Gomez-Lopez, G.; et al. Genomic profiling of patient-derived xenografts for lung cancer identifies B2M inactivation impairing immunorecognition. Clin. Cancer Res. 2016, 23, 3203–3213. [Google Scholar] [CrossRef] [Green Version]
  98. Gettinger, S.; Choi, J.; Hastings, K.; Truini, A.; Datar, I.; Sowell, R.; Wurtz, A.; Dong, W.; Cai, G.; Melnick, M.A.; et al. Impaired HLA class I antigen processing and presentation as a mechanism of acquired resistance to immune checkpoint inhibitors in lung cancer. Cancer Discov. 2017, 7, 1420–1435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Perea, F.; Bernal, M.; Sanchez-Palencia, A.; Carretero, J.; Torres, C.; Bayarri, C.; Gomez-Morales, M.; Garrido, F.; Ruiz-Cabello, F. The absence of HLA class I expression in non-small cell lung cancer correlates with the tumor tissue structure and the pattern of T cell infiltration. Int. J. Cancer 2017, 140, 888–899. [Google Scholar] [CrossRef]
  100. Burr, M.L.; Sparbier, C.E.; Chan, K.L.; Chan, Y.; Kersbergen, A.; Lam, E.Y.N.; Azidis-Yates, E.; Vassiliadis, D.; Bell, C.C.; Gilan, O.; et al. An evolutionarily conserved function of polycomb silences the MHC class I antigen presentation pathway and enables immune evasion in cancer. Cancer Cell 2019, 36, 385–401. [Google Scholar] [CrossRef] [Green Version]
  101. Lai, Q.; Wang, H.; Li, A.; Xu, Y.; Tang, L.; Chen, Q.; Zhang, C.; Gao, Y.; Song, J.; Du, Z. Decitibine improve the efficiency of anti-PD-1 therapy via activating the response to IFN/PD-L1 signal of lung cancer cells. Oncogene 2018, 37, 2302–2312. [Google Scholar] [CrossRef]
  102. Motz, G.T.; Santoro, S.P.; Wang, L.; Garrabrant, T.; Lastra, R.R.; Hagemann, I.S.; Lal, P.; Feldman, M.D.; Benencia, F.; Coukos, G. Tumor endothelium FasL establishes a selective immune barrier promoting tolerance in tumors. Nat. Med. 2014, 20, 607–615. [Google Scholar] [CrossRef] [PubMed]
  103. Tessema, M.; Klinge, D.M.; Yingling, C.M.; Do, K.; van Neste, L.; Belinsky, S.A. Re-expression of CXCL14, a common target for epigenetic silencing in lung cancer, induces tumor necrosis. Oncogene 2010, 29, 5159–5170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. 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] [Green Version]
  105. Molon, B.; Ugel, S.; Del Pozzo, F.; Soldani, C.; Zilio, S.; Avella, D.; De Palma, A.; Mauri, P.; Monegal, A.; Rescigno, M.; et al. Chemokine nitration prevents intratumoral infiltration of antigen-specific T cells. J. Exp. Med. 2011, 208, 1949–1962. [Google Scholar] [CrossRef]
  106. Chung, L.; Tang, S.; Sun, G.; Chou, T.; Yeh, T.; Yu, S.; Sun, K. Galectin-1 promotes lung cancer progression and chemoresistance by upregulating p38 MAPK, ERK, and cyclooxygenase-2. Clin. Cancer Res. 2012, 18, 4037–4047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Nambiar, D.K.; Aguilera, T.; Cao, H.; Kwok, S.; Kong, C.; Bloomstein, J.; Wang, Z.; Rangan, V.S.; Jiang, D.; von Eyben, R.; et al. Galectin-1-driven T cell exclusion in the tumor endothelium promotes immunotherapy resistance. J. Clin. Investig. 2019, 129, 5553–5567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Kowanetz, M.; Zou, W.; Gettinger, S.N.; Koeppen, H.; Kockx, M.; Schmid, P.; Kadel, E.E., III; Wistuba, I.; Chaft, J.; Rizvi, N.A.; et al. Differential regulation of PD-L1 expression by immune and tumor cells in NSCLC and the response to treatment with atezolizumab (anti-PD-L1). Proc. Natl. Acad. Sci. USA 2018, 115, E10119–E10126. [Google Scholar] [CrossRef] [Green Version]
  109. 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. 2015, 6, 202–216. [Google Scholar] [CrossRef] [Green Version]
  110. 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] [Green Version]
  111. Frank, R.; Scheffler, M.; Merkelbach-Bruse, S.; Ihle, M.A.; Kron, A.; Rauer, M.; Ueckeroth, F.; König, K.; Michels, S.; Fischer, R.; et al. Clinical and pathological characteristics of KEAP1- and NFE2L2-mutated non-small cell lung carcinoma (NSCLC). Clin. Cancer Res. 2018, 24, 3087–3096. [Google Scholar] [CrossRef] [Green Version]
  112. Nadal, E.; Palmero, R.; Muñoz-Pinedo, C. Mutations in the antioxidant KEAP1/NRF2 pathway define an aggressive subset of NSCLC resistant to conventional treatments. J. Thorac. Oncol. 2019, 14, 1881–1883. [Google Scholar] [CrossRef] [PubMed]
  113. Truica, M.I.; Burns, M.C.; Han, H.; Abdulkadir, S.A. Turning up the heat on Myc: Progress in small-molecule inhibitors. Cancer Res. 2021, 81. in press. [Google Scholar] [CrossRef] [PubMed]
  114. Sampson, J.H.; Gunn, M.D.; Fecci, P.E.; Ashley, D.M. Brain immunology and immunotherapy in brain tumours. Nat. Rev. Cancer 2020, 20, 12–25. [Google Scholar] [CrossRef] [PubMed]
  115. Rodriguez, E.F.; De Marchi, F.; Lokhandwala, P.M.; Belchis, D.; Xian, R.; Gocke, C.D.; Eshleman, J.R.; Illei, P.; Li, M. IDH1 and IDH2 mutations in lung adenocarcinomas: Evidences of subclonal evolution. Cancer Med. 2020, 9, 4386–4394. [Google Scholar] [CrossRef] [Green Version]
  116. 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]
  117. Schoenfeld, A.J.; Bandlamudi, C.; Lavery, J.A.; Montecalvo, J.; Namakydoust, A.; Rizvi, H.; Egger, J.; Concepcion, C.P.; Paul, S.; Arcila, M.E.; et al. The genomic landscape of SMARCA4 alterations and associations with outcomes in patients with lung cancer. Clin. Cancer. Res. 2020, 26, 5701–5708. [Google Scholar] [CrossRef]
  118. Biton, J.; Mansuet-Lupo, A.; Pécuchet, N.; Alifano, M.; Ouakrim, H.; Arrondeau, J.; Boudou-Rouquette, P.; Goldwasser, F.; Leroy, K.; Goc, J.; et al. TP53, STK11, and EGFR mutations predict tumor immune profile and the response to anti-PD-1 in lung adenocarcinoma. Clin. Cancer Res. 2018, 24, 5710–5723. [Google Scholar] [CrossRef] [Green Version]
  119. Liu, C.; Zheng, S.; Jin, R.; Wang, X.; Wang, F.; Zang, R.; Xu, H.; Lu, Z.; Huang, J.; Lei, Y.; et al. The superior efficacy of anti-PD-1/PD-L1 immunotherapy in KRAS-mutant non-small cell lung cancer that correlates with an inflammatory phenotype and increased immunogenicity. Cancer Lett. 2020, 470, 95–105. [Google Scholar] [CrossRef]
  120. Jeanson, A.; Tomasini, P.; Souquet-Bressand, M.; Brandone, N.; Boucekine, M.; Grangeon, M.; Chaleat, S.; Khobta, N.; Milia, J.; Mhanna, L.; et al. Efficacy of immune checkpoint inhibitors in KRAS-mutant non-small cell lung cancer (NSCLC). J. Thorac. Oncol. 2019, 14, 1095–1101. [Google Scholar] [CrossRef]
  121. Hirsch, F.R.; Suda, K.; Wiens, J.; Bunn, P.A., Jr. New and emerging targeted treatments in advanced non-small-cell lung cancer. Lancet 2016, 388, 1012–1024. [Google Scholar] [CrossRef]
  122. Swanton, C.; Govindan, R. Clinical implications of genomic discoveries in lung cancer. N. Engl. J. Med. 2016, 374, 1864–1873. [Google Scholar] [CrossRef] [Green Version]
  123. Zhang, Y.; Guan, X.; Jiang, P. Cytokine and chemokine signals of T-cell exclusion in tumors. Front. Immunol. 2020, 11, 594609. [Google Scholar] [CrossRef]
  124. Frezzetti, D.; Gallo, M.; Maiello, M.R.; D’Alessio, A.; Esposito, C.; Chicchinelli, N.; Normanno, N.; De Luca, A. VEGF as a potential target in lung cancer. Expert Opin. Ther. Targets 2017, 21, 959–966. [Google Scholar] [CrossRef] [PubMed]
  125. Toonkel, R.L.; Borczuk, A.C.; Powell, C.A. TGF-β signaling pathway in lung adenocarcinoma invasion. J. Thorac. Oncol. 2010, 5, 153–157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Voron, T.; Colussi, O.; Marcheteau, E.; Pernot, S.; Nizard, M.; Pointet, A.; Latreche, S.; Bergaya, S.; Benhamouda, N.; Tanchot, C.; et al. VEGF-A modulates expression of inhibitory checkpoints on CD8+ T cells in tumors. J. Exp. Med. 2015, 212, 139–148. [Google Scholar] [CrossRef] [PubMed]
  127. Huang, H.; Langenkamp, E.; Georganaki, M.; Loskog, A.; Fuchs, P.F.; Dieterich, L.C.; Kreuger, J.; Dimberg, A. VEGF suppresses T-lymphocyte infiltration in the tumor microenvironment through inhibition of NK-κB-induced endothelial activation. FASEB J. 2015, 29, 227–238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Wu, X.; Giobbie-Hurder, A.; Liao, X.; Lawrence, D.; McDermott, D.; Zhou, J.; Rodig, S.; Hodi, F.S. VEGF neutralization plus CTLA-4 blockade alters soluble and cellular factors associated with enhancing lymphocyte infiltration and humoral recognition in melanoma. Cancer Immunol. Res. 2016, 4, 858–868. [Google Scholar] [CrossRef] [Green Version]
  129. Yang, J.; Yan, J.; Liu, B. Targeting VEGF/VEGFR to modulate antitumor immunity. Front. Immunol. 2018, 9, 978. [Google Scholar] [CrossRef] [Green Version]
  130. Allen, E.; Jabouille, A.; Rivera, L.B.; Lodewijckx, I.; Missiaen, R.; Steri, V.; Feyen, K.; Tawney, J.; Hanahan, D. Combined antiangiogenic and anti-PD-L1 therapy stimulates tumor immunity through HEV formation. Sci. Transl. Med. 2017, 9, eaak9679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Socinski, M.A.; Jotte, R.M.; Cappuzzo, F.; Orlandi, F.; Stroyakovskiy, D.; Nogami, N.; Rodríguez-Abreu, D.; Moro-Sibilot, D.; Thomas, C.A.; Barlesi, F.; et al. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N. Engl. J. Med. 2018, 378, 2288–2301. [Google Scholar] [CrossRef]
  132. Bierie, B.; Moses, H.L. Transforming growth factor beta (TGF-β) and inflammation in cancer. Cytokine Growth Factor Rev. 2010, 21, 49–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. 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 reconstructed colon cancer metastasis. Nature 2018, 554, 538–543. [Google Scholar] [CrossRef] [Green Version]
  134. 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. TGF-β attenuates tumor response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018, 554, 544–548. [Google Scholar] [CrossRef] [PubMed]
  135. Takasaka, N.; Seed, R.I.; Cormier, A.; Bondesson, A.J.; Lou, J.; Elattma, A.; Ito, S.; Yanagisawa, H.; Hashimoto, M.; Ma, R.; et al. Integrin αvβ8-expressing tumor cells evade host immunity by regulating TGF-β activation in immune cells. JCI Insight 2018, 3, e122591. [Google Scholar] [CrossRef] [Green Version]
  136. Yang, L.; Pang, Y.; Moses, H.L. TGF-β and immune cells: An important regulatory axis in the tumor microenvironment and progression. Trends Immunol. 2010, 31, 220–227. [Google Scholar] [CrossRef] [Green Version]
  137. Holmgaard, R.B.; Schaer, D.A.; Li, Y.; Castaneda, S.P.; Murphy, M.Y.; Xu, X.; Inigo, I.; Dobkin, J.; Manro, J.R.; Iversen, P.W.; et al. Targeting the TGFβ pathway with galunisertib, a TGFβRI small molecule inhibitor, promotes anti-tumor immunity leading to durable, complete responses, as monotherapy and in combination with checkpoint blockade. J. Immunother. Cancer 2018, 6, 47. [Google Scholar] [CrossRef]
  138. Demaria, S.; Coleman, C.N.; Formenti, S.C. Radiotherapy: Changing the game in immunotherapy. Trends Cancer 2016, 2, 286–294. [Google Scholar] [CrossRef] [Green Version]
  139. Durante, M.; Brenner, D.J.; Formenti, S.C. Does heavy ion therapy work through the immune system? Int. J. Radiat. Oncol. Biol. Phys. 2016, 96, 934–936. [Google Scholar] [CrossRef]
  140. Heinhuis, K.M.; Ros, W.; Kok, M.; Steeghs, N.; Beijnen, J.H.; Schellens, J.H.M. Enhancing antitumor response by combining immune checkpoint inhibitors with chemotherapy in solid tumors. Ann. Oncol. 2019, 30, 219–235. [Google Scholar] [CrossRef] [PubMed]
  141. Rodríguez-Ruiz, M.E.; Vanpouille-Box, C.; Melero, I.; Formenti, S.C.; Demaria, S. Immunological mechanisms responsible for radiation-induced abscopal effect. Trends Immunol. 2018, 39, 644–655. [Google Scholar] [CrossRef]
  142. Schadt, L.; Sparano, C.; Schweiger, N.A.; Silina, K.; Cecconi, V.; Lucchiari, G.; Yagita, H.; Guggisberg, E.; Saba, S.; Nascakova, Z.; et al. Cancer-cell-intrinsic cGAS expression mediates tumor immunogenicity. Cell Rep. 2019, 29, 1236–1248. [Google Scholar] [CrossRef] [Green Version]
  143. Kalbasi, A.; Tariveranmoshabad, M.; Hakimi, K.; Kremer, S.; Campbell, K.M.; Funes, J.M.; Vega-Crespo, A.; Parisi, G.; Champekar, A.; Nguyen, C.; et al. Uncoupling interferon signaling and antigen presentation to overcome immunotherapy resistance due to JAK1 loss in melanoma. Sci. Transl. Med. 2020, 12, eabb0152. [Google Scholar] [CrossRef]
  144. Yamamoto, T.N.; Lee, P.; Vodnala, S.K.; Gurusamy, D.; Kishton, R.J.; Yu, Z.; Eidizadeh, A.; Eil, R.; Fioravanti, J.; Gattinoni, L.; et al. T cells genetically engineered to overcome death signaling enhance adoptive cancer immunotherapy. J. Clin. Investig. 2019, 129, 1551–1565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Ngiow, S.F.; von Scheidt, B.; Akiba, H.; Yagita, H.; Teng, M.W.L.; Smyth, M.J. Anti-TIM3 antibody promotes T cell IFN-γ-mediated antitumor immunity and suppresses established tumors. Cancer Res. 2011, 71, 3540–3551. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Burova, E.; Hermann, A.; Dai, J.; Ullman, E.; Halasz, G.; Potocky, T.; Hong, S.; Liu, M.; Allbritton, O.; Woodruff, A.; et al. Preclinical development of the anti-LAG-3 antibody REGN3767: Characterization and activity in combination with the anti-PD-1 antibody cemiplimab in human PD-1xLAG-3-knockin mice. Mol. Cancer Ther. 2019, 18, 2051–2062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Preillon, J.; Cuende, J.; Rabolli, V.; Garnero, L.; Mercier, M.; Wald, N.; Pappalardo, A.; Denies, S.; Jamart, D.; Michaux, A.; et al. Restoration of T-cell effector function, depletion of Tregs, and direct killing of tumor cells: The multiple mechanisms of action of a-TIGIT antagonist antibodies. Mol. Cancer Ther. 2021, 20, 121–131. [Google Scholar] [CrossRef] [PubMed]
  148. Chen, Y.; Lin, H.; Chien, C.; Lai, Y.; Sun, W.; Chen, C.; Cheng, W. BTLA blockade enhances cancer therapy by inhibiting IL-6/IL-10-induced CD19high B lymphocytes. J. Immunother. Cancer 2019, 7, 313. [Google Scholar] [CrossRef] [PubMed]
  149. Pfannenstiel, L.W.; Diaz-Montero, C.M.; Tian, Y.F.; Scharpf, J.; Ko, J.S.; Gastman, B.R. Immune-checkpoint blockade opposes CD8+ T-cell suppression in human and murine cancer. Cancer Immunol. Res. 2019, 7, 510–525. [Google Scholar] [CrossRef] [Green Version]
  150. Martinez, M.; Kim, S.; St. Jean, N.; O’Brien, S.; Lian, L.; Sun, J.; Verona, R.I.; Moon, E. Addition of anti-TIM3 or anti-TIGIT antibodies to anti-PD1 blockade augments human T cell adoptive cell transfer. Oncoimmunology 2021, 10, 1873607. [Google Scholar] [CrossRef]
  151. Grapin, M.; Richard, C.; Limagne, E.; Boidot, R.; Morgand, V.; Bertaut, A.; Derangere, V.; Laurent, P.; Thibaudin, M.; Fumet, J.D.; et al. Optimized fractionated radiotherapy with anti-PD-L1 and anti-TIGIT: A promising new combination. J. Immunother. Cancer 2019, 9, 160. [Google Scholar] [CrossRef] [Green Version]
  152. Ciardiello, D.; Elez, E.; Tabernero, J.; Seoane, J. Clinical development of therapies targeting TGFβ: Current knowledge and future perspectives. Ann. Oncol. 2020, 31, 1336–1349. [Google Scholar] [CrossRef]
  153. David, J.M.; Dominguez, C.; McCampbell, K.K.; Gulley, J.L.; Schlom, J.; Palena, C. A novel bifunctional anti-PD-L1/TGF-β trap fusion protein (M7824) efficiently reverts mesenchymalization of human lung cancer cells. OncoImmunology 2017, 6, e1349589. [Google Scholar] [CrossRef] [Green Version]
  154. Prendergast, G.C.; Malachowski, W.P.; DuHadaway, J.B.; Muller, A.J. Discovery of IDO1 inhibitors: From bench to bedside. Cancer Res. 2017, 77, 6795–6811. [Google Scholar] [CrossRef] [Green Version]
  155. Onda, M.; Kobayashi, K.; Pastan, I. Depletion of regulatory T cells in tumors with an anti-CD25 immunotoxin induces CD8 T cell-mediated systemic antitumor immunity. Proc. Natl. Acad. Sci. USA 2019, 116, 4575–4582. [Google Scholar] [CrossRef] [Green Version]
  156. Kim, B.; Clinton, J.; Wang, Q.; Chang, S.H. Targeting ST2 expressing activated regulatory T cells in Kras-mutant lung cancer. Oncoimmunology 2020, 9, e1682380. [Google Scholar] [CrossRef] [Green Version]
  157. Ji, D.; Song, C.; Li, Y.; Xia, J.; Wu, Y.; Jia, J.; Cui, X.; Yu, S.; Gu, J. Combination of radiotherapy and suppression of Tregs enhances abscopal antitumor effect and inhibits metastasis in rectal cancer. J. Immunother. Cancer. 2020, 8, e000826. [Google Scholar] [CrossRef] [PubMed]
  158. Hashimoto, A.; Fukumoto, T.; Zhang, R.; Gabrilovich, D. Selective targeting of different populations of myeloid-derived suppressor cells by histone deacetylase inhibitors. Cancer Immunol. Immunother. 2020, 69, 1929–1936. [Google Scholar] [CrossRef] [PubMed]
  159. Fultang, L.; Panetti, S.; Ng, M.; Collins, P.; Graef, S.; Rizkalla, N.; Booth, S.; Lenton, R.; Noyvert, B.; Shannon-Lowe, C.; et al. MDSC targeting with Gemtuzumab ozogammicin restores T cell immunity and immunotherapy against cancer. EBioMedicine 2019, 47, 235–246. [Google Scholar] [CrossRef] [Green Version]
  160. Greene, S.; Robbins, Y.; Mydlarz, W.K.; Huynh, A.P.; Schmitt, N.C.; Friedman, J.; Horn, L.A.; Palena, C.; Schlom, J.; Maeda, D.Y.; et al. Inhibition of MDSC trafficking with SX-682, a CXCR1/2 inhibitor, enhances NK-cell immunotherapy in head and neck cancer models. Clin. Cancer Res. 2020, 26, 1420–1431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Grimaldi, A.; Cammarata, I.; Martire, C.; Focaccetti, C.; Piconese, S.; Buccilli, M.; Mancone, C.; Buzzacchino, F.; Berrios, J.R.G.; D’Alessandris, N.; et al. Combination of chemotherapy and PD-1 blockade induces T cell responses to tumor non-mutated neoantigens. Commun. Biol. 2020, 3, 85. [Google Scholar] [CrossRef] [Green Version]
  162. Demaria, S.; Golden, E.B.; Formenti, S.C. Role of local radiation therapy in cancer immunotherapy. JAMA Oncol. 2015, 1, 1325–1332. [Google Scholar] [CrossRef]
  163. Wan, S.; Pestka, S.; Jubin, R.G.; Lyu, Y.L.; Tsai, Y.; Liu, L.F. Chemotherapeutics and radiation stimulate MHC class I expression through elevated interferon-beta signaling in breast cancer cells. PLoS ONE 2012, 7, e32542. [Google Scholar]
  164. Reits, E.A.; Hodge, J.W.; Herberts, C.A.; Groothuis, T.A. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J. Exp. Med. 2006, 203, 1259–1271. [Google Scholar] [CrossRef]
  165. Zebertavage, L.K.; Alice, A.; Crittenden, M.R.; Gough, M.J. Transcriptional upregulation of NLRC5 by radiation drives STING- and interferon-independent MHC-I expression on cancer cells and T cell cytotoxicity. Sci. Rep. 2020, 10, 7376. [Google Scholar] [CrossRef]
  166. Herter-Sprie, G.S.; Koyama, S.; Korideck, H.; Hai, J.; Deng, J.; Li, Y.Y.; Buczkowski, K.A.; Grant, A.K.; Ullas, S.; Rhee, K.; et al. Synergy of radiotherapy and PD-1 blockade in Kras-mutant lung cancer. JCI Insight 2016, 1, e87415. [Google Scholar] [CrossRef]
  167. Dovedi, S.J.; Cheadle, E.J.; Popple, A.L.; Poon, E.; Morrow, M.; Stewart, R.; Yusko, E.C.; Sanders, C.M.; Vignali, M.; Emerson, R.O.; et al. Fractionated radiation therapy stimulates anti-tumor immunity mediated by both resident and infiltrating polyclonal T-cell populations when combined with PD1 blockade. Clin. Cancer Res. 2017, 23, 5514–5526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Formenti, S.C.; Rudqvist, N.; Golden, E.; Cooper, B.; Wennerberg, E.; Lhuillier, C.; Vanpouille-Box, C.; Friedman, K.; de Andrade, L.F.; Wucherpfennig, K.W.; et al. Radiotherapy induces responses of lung cancer to CTLA-4 blockade. Nat. Med. 2018, 24, 1845–1851. [Google Scholar] [CrossRef] [PubMed]
  169. Antonia, S.J.; Villegas, A.; Daniel, D.; Vicente, D.; Murakami, S.; Hui, R.; Kurata, T.; Chiappori, A.; Lee, K.H.; de Wit, M.; et al. Overall survival with durvalumab after chemoradiotherapy in stage III NSCLC. N. Engl. J. Med. 2018, 379, 2342–2350. [Google Scholar] [CrossRef] [PubMed]
  170. Lind, H.; Gameiro, S.R.; Jochems, C.; Donahue, R.N.; Strauss, J.; Gulley, J.L.; Palena, C.; Schlom, J. Dual targeting of TGF-β and PD-L1 via a bifunctional anti-PD-L1/ TGF-βRII agent: Status of preclinical and clinical advances. J. Immunother. Cancer 2020, 8, e000433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Li, L.; Wei, J.; Dong, J.; Lin, Q.; Tang, H.; Jia, Y.; Tan, W.; Chen, Q.; Zeng, T.; Xing, S.; et al. Laminin γ2-mediating T cell exclusion attenuates response to anti-PD-1 therapy. Sci. Adv. 2021, 7, eabc8346. [Google Scholar] [CrossRef]
  172. Alexandrov, L.B.; Nik-Zainal, S.; Wedge, D.C.; Aparicio, S.A.J.R.; Behjati, S.; Biankin, A.V.; Bignell, G.R.; Bolli, N.; Borg, A.; Børresen-Dale, A.; et al. Signatures of mutational processes in human cancer. Nature 2013, 500, 415–421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Le, X.; Negrao, M.V.; Reuben, A.; Federico, L.; Diao, L.; McGrail, D.; Nilsson, M.; Robichaux, J.; Munoz, I.G.; Patel, S.; et al. Characterization of the immune landscape of EGFR-mutant NSCLC identifies CD73/adenosine pathway as a potential therapeutic target. J. Thorac. Oncol. 2021, 16, 583–600. [Google Scholar] [CrossRef] [PubMed]
  174. Mazieres, J.; Drilon, A.; Lusque, A.; Mhanna, L.; Cortot, A.B.; Mezquita, L.; Thai, A.A.; Mascaux, C.; Couraud, S.; Veillon, R.; et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: Results from the IMMUNOTARGET registry. Ann. Oncol. 2019, 30, 1321–1328. [Google Scholar] [CrossRef]
  175. Offin, M.; Guo, R.; Wu, S.L.; Sabari, J.; Land, J.D.; Ni, A.; Montecalvo, J.; Halpenny, D.F.; Buie, L.W.; Pak, T.; et al. Immunophenotype and response to immunotherapy of RET-rearranged lung cancers. JCO Precis. Oncol. 2019, 3, PO.18.00386. [Google Scholar] [CrossRef] [PubMed]
  176. Guisier, F.; Dubos-Arvis, C.; Viñas, F.; Doubre, H.; Ricordel, C.; Ropert, S.; Janicot, H.; Bernardi, M.; Fournel, P.; Lamy, R.; et al. Efficacy and safety of anti-PD-1 immunotherapy in patients with advanced NSCLC with BRAF, HER2, or MET mutations or RET translocation: GFPC 01-2018. J. Thorac. Oncol. 2020, 15, 628–636. [Google Scholar] [CrossRef]
  177. Skoulidis, F.; Heymach, J.V. Co-occurring genomic alterations in non-small-cell lung cancer biology and therapy. Nat. Rev. Cancer 2019, 19, 495–509. [Google Scholar] [CrossRef]
  178. Skoulidis, F.; Goldberg, M.E.; Greenawalt, D.M.; Hellmann, M.D.; Awad, M.M.; Gainor, J.F.; Schrock, A.B.; Hartmaier, R.J.; Trabucco, S.E.; Gay, L.; et al. STK11/LKB1 mutations and PD-1 inhibitor resistance in KRAS-mutant lung adenocarcinoma. Cancer Discov. 2018, 8, 822–835. [Google Scholar] [CrossRef] [Green Version]
  179. Kitajima, S.; Ivanova, E.; Guo, S.; Yoshida, R.; Campisi, M.; Sundararaman, S.K.; Tange, S.; Mitsuishi, Y.; Thai, T.C.; Masuda, S.; et al. Suppression of STING associated with LKB1 loss in KRAS-driven lung cancer. Cancer Discov. 2019, 9, 34–45. [Google Scholar] [CrossRef] [Green Version]
  180. Kaufman, J.M.; Amann, J.M.; Park, K.; Arasada, R.R.; Li, H.; Shyr, Y.; Carbone, D.P. LKB1 loss induces characteristic patterns of gene expression in human tumors associated with NRF2 activation and attenuation of PI3K-AKT. J. Thorac. Oncol. 2014, 9, 794–804. [Google Scholar] [CrossRef] [Green Version]
  181. Olagnier, D.; Brandtoft, A.M.; Gunderstofte, C.; Villadsen, N.L.; Krapp, C.; Thielke, A.L.; Laustsen, A.; Peri, S.; Hansen, A.L.; Bonefeld, L.; et al. Nrf2 negatively regulates STING indicating a link between antiviral sensing and metabolic reprogramming. Nat. Comm. 2018, 9, 3506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Goeman, F.; De Nicola, F.; Scalera, S.; Sperati, F.; Gallo, E.; Ciuffreda, L.; Pallocca, M.; Pizzuti, L.; Krasniqi, E.; Barchiesi, G.; et al. Mutations in the KEAP1-NFE2L2 pathway define a molecular subset of rapidly progressing lung adenocarcinoma. J. Thorac. Oncol. 2019, 14, 1924–1934. [Google Scholar] [CrossRef] [PubMed]
  183. Galan-Cobo, A.; Sitthideatphaiboon, P.; Qu, X.; Poteete, A.; Pisegna, M.A.; Tong, P.; Chen, P.; Boroughs, L.K.; Rodriguez, M.L.M.; Zhang, W.; et al. LKB1 and KEAP1/NRF2 pathways cooperatively promote metabolic reprogramming with enhanced glutamine dependence in KRAS-mutant lung adenocarcinoma. Cancer Res. 2019, 79, 3251–3267. [Google Scholar] [CrossRef] [PubMed]
  184. Binkley, M.S.; Jeon, Y.; Nesselbush, M.; Moding, E.J.; Nabet, B.Y.; Almanza, D.; Kunder, C.; Stehr, H.; Yoo, C.H.; Rhee, S.; et al. KEAP1/NFE2L2 mutations predict lung cancer radiation resistance that can be targeted by glutaminase inhibition. Cancer Discov. 2020, 10, 1826–1841. [Google Scholar] [CrossRef] [PubMed]
  185. Sitthideatphaiboon, P.; Galan-Cobo, A.; Negrao, M.V.; Qu, X.; Poteete, A.; Zhang, F.; Liu, D.D.; Lewis, W.E.; Kemp, H.N.; Lewis, J.; et al. STK11/LKB1 mutations in NSCLC are associated with KEAP1/NRF2-dependent radiotherapy resistance targetable by glutaminase inhibition. Clin. Cancer Res. 2021, 27, 1720–1733. [Google Scholar] [CrossRef] [PubMed]
  186. Choi, E.J.; June, B.J.; Lee, S.H.; Yoo, H.S.; Shin, E.A.; Ko, H.J.; Chang, S.; Kim, S.Y.; Jeon, S.M. A clinical drug library screen identifies clobetasol propionate as an NRF2 inhibitor with potential therapeutic efficacy in KEAP1 mutant lung cancer. Oncogene 2017, 36, 5285–5295. [Google Scholar] [CrossRef]
  187. Stakheev, D.; Taborska, P.; Strizova, Z.; Podrazil, M.; Bartunkova, J.; Smrz, D. The Wnt/β-catenin signaling inhibitor XAV939 enhances the elimination of LNCaP and PC-3 prostate cancer cells by prostate cancer patient lymphocytes in vitro. Sci. Rep. 2019, 9, 4761. [Google Scholar] [CrossRef]
  188. Vidotto, T.; Melo, C.M.; Castelli, E.; Koti, M.; dos Reis, R.B.; Squire, J.A. Emerging role of PTEN loss in evasion of the immune response to tumours. Br. J. Cancer 2020, 122, 1732–1743. [Google Scholar] [CrossRef]
  189. Chaft, J.E.; Arcila, M.E.; Paik, P.K.; Lau, C.; Riely, G.J.; Pietanza, M.C.; Zakowski, M.F.; Rusch, V.; Sima, C.S.; Ladanyi, M.; et al. Coexistence of PIK3CA and other oncogene mutations in lung adenocarcinoma-rationale for comprehensive mutation profiling. Mol. Cancer Ther. 2011, 11, 485–491. [Google Scholar] [CrossRef] [Green Version]
  190. Dong, Z.Y.; Zhang, J.T.; Liu, S.Y.; Su, J.; Zhang, C.; Xie, Z.; Zhou, Q.; Tu, H.Y.; Xu, C.R.; Yan, L.X.; et al. EGFR mutation correlates with uninflamed phenotype and weak immunogenicity, causing impaired response to PD-1 blockade in non-small cell lung cancer. Oncoimmunology 2017, 6, e1356145. [Google Scholar] [CrossRef] [Green Version]
  191. Lisberg, A.; Cummings, A.; Goldman, J.W.; Bornazyan, K.; Reese, N.; Wang, T.; Coluzzi, P.; Ledezma, B.; Mendenhall, M.; Hunt, J.; et al. A phase II study of pembrolizumab in EGFR-mutant, PD-L1+, tyrosine kinase inhibitor naïve patients with advanced NSCLC. J. Thorac. Oncol. 2018, 13, 1138–1145. [Google Scholar] [CrossRef] [Green Version]
  192. Hastings, K.; Yu, H.A.; Wei, W.; Sanchez-Vega, F.; DeVeaux, M.; Choi, J.; Rizvi, H.; Lisberg, A.; Truini, A.; Lydon, C.A.; et al. EGFR mutation subtypes and response to immune checkpoint blockade treatment in non-small-cell lung cancer. Ann. Oncol. 2019, 30, 1311–1320. [Google Scholar] [CrossRef] [Green Version]
  193. Pyo, K.; Lim, S.M.; Park, C.; Jo, H.; Kim, J.H.; Yun, M.; Kim, D.; Xin, C.; Lee, W.; Gheorghiu, B.; et al. Comprehensive analyses of immunodynamics and immunoreactivity in response to treatment in ALK-positive non-small-cell lung cancer. J. Immunother. Cancer 2020, 8, e000970. [Google Scholar] [CrossRef]
  194. Haratani, K.; Hayashi, H.; Tanaka, T.; Kaneda, H.; Togashi, Y.; Sakai, K.; Hayashi, K.; Tomida, S.; Chiba, Y.; Yonesaka, K.; et al. Tumor immune microenvironment and nivolumab efficacy in EGFR mutation-positive non-small-cell lung cancer based on T790M status after disease progression during EGFR-TKI treatment. Ann. Oncol. 2017, 28, 1532–1539. [Google Scholar] [CrossRef]
  195. Gettinger, S.; Hellmann, M.D.; Chow, L.Q.M.; Borghaei, H.; Antonia, S.; Brahmer, J.R.; Goldman, J.W.; Gerber, D.E.; Juergens, R.A.; Shepherd, F.A.; et al. Nivolumab plus erlotinib in patients with EGFR-mutant advanced NSCLC. J. Thorac. Oncol. 2018, 13, 1363–1372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Schoenfeld, A.J.; Arbour, K.C.; Rizvi, H.; Iqbal, A.N.; Gadgeel, S.M.; Girshman, J.; Kris, M.G.; Riely, G.J.; Yu, H.A.; Hellmann, M.D.; et al. Severe immune-related adverse events are common with sequential PD-(L)1 blockade and Osimertinib. Ann. Oncol. 2019, 30, 839–844. [Google Scholar] [CrossRef]
  197. Oxnard, G.R.; Yang, J.C.; Yu, H.; Kim, S.; Saka, H.; Horn, L.; Goto, K.; Ohe, Y.; Mann, H.; Thress, K.S.; et al. TATTON: A multi-arm, phase Ib trial of osimertinib combined with selumetinib, savolitinib, or durvalumab in EGFR-mutant lung cancer. Ann. Oncol. 2020, 31, 507–516. [Google Scholar] [CrossRef] [Green Version]
  198. Spigel, D.R.; Reynolds, C.; Waterhouse, D.; Garon, E.B.; Chandler, J.; Babu, S.; Thurmes, P.; Spira, A.; Jotte, R.; Zhu, J.; et al. Phase 1/2 study of the safety and tolerability of nivolumab plus crizotinib for the first-line treatment of anaplastic lymphoma kinase translocation-positive advanced non-small cell lung cancer (CheckMate 370). J. Thorac. Oncol. 2018, 13, 682–688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Reck, M.; Mok, T.S.K.; Nishio, M.; Jotte, R.M.; Cappuzzo, F.; Orlandi, F.; Stroyakovskiy, D.; Nogami, N.; Rodríguez-Abreu, D.; Moro-Sibilot, D.; et al. Atezolizumab plus bevacizumab and chemotherapy in non-small-cell lung cancer (Impower150): Key subgroup analyses of patients with EGFR mutations or baseline liver metastases in a randomized, open-label phase 3 trial. Lancet Respir. Med. 2019, 7, 387–401. [Google Scholar] [CrossRef]
Figure 1. Characteristics of different types of TIME related to NSCLC’s response to ICI.
Figure 1. Characteristics of different types of TIME related to NSCLC’s response to ICI.
Cancers 13 02924 g001
Table 1. General classification of the tumor immune micro-environment.
Table 1. General classification of the tumor immune micro-environment.
ReferencesMethodCriteriaTIME ClassificationMajor FeaturesAdditional Features
Herbst et al. [19]IHCPD-L1 expressionResponsiveBefore RxBefore Rx
(TC and IC) Increased PD-L1 expressionIncreased expression of another checkpoint (NSCLC):
CD8+ T cell infiltration (TC, IC)B7-H3, CTLA-4, TIM3, LAG3, IDO1, PD-L2
Decreased CX3CL1; increased CTLA-4
Increased IFN-γ and IFN-γ-inducible genes (e.g., IDO1 and CXCL9)
After RxAfter Rx
Increased PD-L1 expressionIncreased tumor IFN-γ expression
(TC, IC)Gene expression pattern of immune activation:
CD8 and Th1 T cell activationgranzyme-A, B; Perforin, EOMES, IFN-γ, TNF
CXCL10, CD8A, CTLA 4
Non-ResponsivePre-Rx and After RxAfter Rx
Immunological ignoranceLittle or no TILsNo overexpression of genes associated with immune activation
Non-functional immune responseTIL without PD-L1 expressionNo overexpression of genes associated with immune activation
(with pre-treatment CD 8 T cell infiltrate)
Excluded infiltrateImmune infiltrate at tumor marginSame as the two types above, except with increased CTLA-4 expression
Proliferation and PD-L1 expression in immune cells at tumor margin
Teng et al. [24]IHCPD-L1 expression (TC)Type I (adaptive immune resistance)PD-L1 (+), TIL (+)Immunogenic mutations associated with
TILs increased TILs of higher PD-1, CTLA-4 expression
Type II (immune ignorance)PD-L1 (-), TIL (-)No pre-existing T cell infiltration
Type III (intrinsic induction)PD-L1 (+), TIL (-)More common in oncogenic mutation-driven NSCLC
LUAD: PD-L1 expression-associated EGFR mutations
Type IV (tolerance)PD-L1 (-), TIL (+)Increased myeloid cells
Activation of other immune checkpoints and suppressive pathways
Rx: treatment.
Table 2. Characteristics the TCGA TIME subtype classification.
Table 2. Characteristics the TCGA TIME subtype classification.
TIME SubtypesWound Healing ǂIFN-γ DominantInflammatoryLymphocyte DepletedImmunologically QuietTGF-β Dominant
Leukocyte fraction *Intermed.HighIntermed.LowLowHighest
Lymphocyte fraction (25–55%)HighHighestHighIntermed. lowLowestIntermed.
TIL (H and E)HighHighestIntermed. lowLowLowestIntermed.
Immune cell composition
T cells
CD8 T cells (<15%)Intermed. highHighestHighIntermed. lowLowestIntermed.
CD4 T cells (<35%)
Th1Lowest Elevated ElevatedElevated
Th2HighestHighestLowestIntermed.LowIntermed. high
Tfh (<10%)HighHighestIntermed.LowLowestIntermed. low
Tregs (<5%)HighHighestIntermed. highLowLowestHigh
Macrophages (38–60%) ElevatedMost elevatedElevated
M0 (<15%)HighestHighIntermed. lowIntermed.LowestHigh
M1 (<10%)Intermed.HighestIntermed.Intermed. lowLowestIntermed.
M2 (>20%)Intermed. lowLowestIntermed.HighHighestHigh
Tumor proliferation rateHighestHighestLowHighLowestHigh
Survival
OSIntermediateIntermediateBestWorstWorseWorst
PFIIntermediateIntermediateBestWorstWorseWorst
NSCLC subtypePredom. in LUSC; third common in LUAD **Second most common in LUAD and LUSCPredom. In LUAD ***LUSC **
Factors of immunogenecity
DNA damage
Tumor neoantigen load
SNVsHighestSecond highest Lowest
IndelsHighestSecond highest Lowest
ITHElevatedElevatedLowest
Enriched oncogenic driver mutationsAPC, JAK1, PIK3CA, FGFR3PIK3CA, FGFR3CDH1, PIK3CA, FGFR3EGFR
TCR diversityIntermediateHighestIntermediateLowLowestHighest
Immunomodulators
Expression
CXCL10 Highest LowestSecond Highest
EDNRBLowLowest Highest
BTLA HighHigh
Networks modulating the immune response
Predominant immune cells CD8 T cellsCD8 T cells, CD4 T cellsCD4 T cells CD4 T cells
Intracellular regulatory networks
TGF-β (somatic mut+) ↓Leuk Fract.↑Leuk Fract. ↓Leuk Fract.
r DC, M0, M1, M2, r NK, plasma cells↑E, a Mast, M0/2, a DC, r NK, TγΔ↑M1, M2, N, CD4, Treg↑M0,M1, a DC↑M0, Treg, mr CD4r DC
a NK, Treg, Tfh, CD8↓CD8, Treg, Tfh, a NK↓DC, M0, Tfh, m B cells, plamsa cells↓monocytesn CD4, CD8
Extracellular comm. networks
IFN-γ (+)IFN-γ (+)
TGF-β (+)TGF-β, TGF-βR(+) TGF-β, TGF-βR(+)
T cell and macrophage-related signalingCD80-CTLA4LAG-3, CD27/28CD27, PD-1TLR4, VEGFBTLR4TLR4
CD70-CD27TIGIT, ICOS, CTLA, PD-1CCR4, 5; CXCR3 DARC EDN3-EDNRB, CX3CL1-CX3CR1ITGB2
IL1A/1B-IL1R2CXCR3, CCR1,4,5 CD276
CXCL9-CXCR3BTLA
ǂ associated with high expression of angiogenic genes; a: activated; r: resting; m: mature; n: naïve; * highest in LUSC and LUAD (median: ≈30%); ** decreased survival; *** increased survival; ↑: increased; ↓: decreased.
Table 3. Mutations associated with the most common neoantigens and enriched in different TIME subtypes based on TCGA data.
Table 3. Mutations associated with the most common neoantigens and enriched in different TIME subtypes based on TCGA data.
TIME SubtypeNeoantigen-Related Driver MutationsEnrichment
Wound healingKRAS, KRAS G12, PIKC3A, TP53APC (OM), JAK1 (OM), TP53 *, FAT1, PPP2R1A, BRCA1, RB1, PIK3CA (OM), PTPRD, SPTA1, CTNNB1 *, FGFR3 * (OM), SMARCA4, KRAS G12, DACH1, PTEN *, SMARCA1, JAK1, KRAS *, MSH3
IFN-γ-dominantPIKC3A, TP53CASP8, HLA-A, HLA-B, ZNF750, TP53 *, MLH1, NF1 *, FAT1, PPP2R1A, BRCA1, RB1 *, PIK3CA(OM), PTPRD, SPTA1, DACH1
InflammatoryBRAFBRAF, CDH1 (OM), PBRM1 *
Lymphocyte-depletedIDH1EGFR (OM), CTNNB1 *
Immunologically quietTP53, IDH1IDH1 R132H, ATRX, CIC *, TP53 *
TGF-β-dominantKRAS G12KRAS G12
Bolded: associated with increased leuk. fraction; italics: associated with decreased leuk. fraction; *: associated with expression of known immunomodulators.
Table 4. Potential treatment strategies to enhance un-inflamed NSCLC’s response to ICI.
Table 4. Potential treatment strategies to enhance un-inflamed NSCLC’s response to ICI.
TIME ClassificationCharacteristics/AlterationsProposed Treatment
ToleranceJAK mutationsICI + RT, chemotherapy, STING agonists, dsDNA sensing nano-therapy, and/or adoptive T cell transfer
↑TregsICI + Treg suppressors (e.g., anti-CD25, anti-ST2) ± RT
↑MDSCsICI + MDSC suppressors (targeted therapies, HDAC inhibitors, CXCR1/2 inhibitors, etc.)
↑FasL (MDSC)ICI + adoptive T cell therapy or antibodies to FasL
↑VEGFICI + VEGF inhibitors
↑TGF-βICI + TGF-β inhibitors, fused anti-PD-L1/TGF-β trap
↑IDO1ICI + IDO1 inhibitors
Terminal T cell exhaustion through other immune checkpointsICI of multiple/or alternative immune checkpoints, such as LAG-3, TIM-3, TIGIT, and BTLA ± RT ± chemotherapy
Immunological Ignorance/Exclusion
Lack of TAAs/immuno-editingICI + RT and/or chemotherapy
STK11/LKB1, KEAP1 mutationsICI + glutaminase inhibitors and/or NRF2 inhibitors ± RT, chemotherapy, or other STING activators
Wnt/β-catenin mutationsWnt/β-catenin inhibitors
PTEN loss/PIK3CA mutationsPIK3CA inhibitors
EGFR mutation (exon 21)EGFR TKI with ICI at progression; ICI/chemo/inhibitors of other targets/RT combinations
EGFR mutation (other)ICI/chemo/inhibitors of other targets/RT combinations
ALK or RET re-arrangedICI/chemo/inhibitors of other targets/RT combinations
↑AngiogenesisICI + VEGF inhibitors, COX-2 inhibitors, or FasL antibodies ± chemotherapy
↑TGF-βAs above
TIME: tumor immune micro-environment; ↑: increased (in quantity, expression, or signaling).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Chi, A.; He, X.; Hou, L.; Nguyen, N.P.; Zhu, G.; Cameron, R.B.; Lee, J.M. Classification of Non-Small Cell Lung Cancer’s Tumor Immune Micro-Environment and Strategies to Augment Its Response to Immune Checkpoint Blockade. Cancers 2021, 13, 2924. https://doi.org/10.3390/cancers13122924

AMA Style

Chi A, He X, Hou L, Nguyen NP, Zhu G, Cameron RB, Lee JM. Classification of Non-Small Cell Lung Cancer’s Tumor Immune Micro-Environment and Strategies to Augment Its Response to Immune Checkpoint Blockade. Cancers. 2021; 13(12):2924. https://doi.org/10.3390/cancers13122924

Chicago/Turabian Style

Chi, Alexander, Xia He, Lin Hou, Nam P. Nguyen, Guangying Zhu, Robert B. Cameron, and Jay M. Lee. 2021. "Classification of Non-Small Cell Lung Cancer’s Tumor Immune Micro-Environment and Strategies to Augment Its Response to Immune Checkpoint Blockade" Cancers 13, no. 12: 2924. https://doi.org/10.3390/cancers13122924

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