Next Article in Journal
Changes in the Ultrastructure of the Bladder Urothelium in Patients with Interstitial Cystitis after Intravesical Injections of Platelet-Rich Plasma
Next Article in Special Issue
Urinary ACE Phenotyping as a Research and Diagnostic Tool: Identification of Sex-Dependent ACE Immunoreactivity
Previous Article in Journal
HDL as Bidirectional Lipid Vectors: Time for New Paradigms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Challenges and the Evolving Landscape of Assessing Blood-Based PD-L1 Expression as a Biomarker for Anti-PD-(L)1 Immunotherapy

Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(5), 1181; https://doi.org/10.3390/biomedicines10051181
Submission received: 29 March 2022 / Revised: 16 May 2022 / Accepted: 17 May 2022 / Published: 20 May 2022

Abstract

:
While promising, PD-L1 expression on tumor tissues as assessed by immunohistochemistry has been shown to be an imperfect biomarker that only applies to a limited number of cancers, whereas many patients with PD-L1-negative tumors still respond to anti-PD-(L)1 immunotherapy. Recent studies using patient blood samples to assess immunotherapeutic responsiveness suggests a promising approach to the identification of novel and/or improved biomarkers for anti-PD-(L)1 immunotherapy. In this review, we discuss the advances in our evolving understanding of the regulation and function of PD-L1 expression, which is the foundation for developing blood-based PD-L1 as a biomarker for anti-PD-(L)1 immunotherapy. We further discuss current knowledge and clinical study results for biomarker identification using PD-L1 expression on tumor and immune cells, exosomes, and soluble forms of PD-L1 in the peripheral blood. Finally, we discuss key challenges for the successful development of the potential use of blood-based PD-L1 as a biomarker for anti-PD-(L)1 immunotherapy.

1. Introduction

Anti-programmed death-1 (PD-1) or anti-programmed death ligand 1 (PD-L1) immunotherapy (anti-PD-(L)1 immunotherapy) has achieved unprecedented clinical efficacy for patients with various types and stages of cancers. The U.S. Food and Drug Administration (FDA) has approved four anti-PD-1 antibodies (nivolumab, pembrolizumab, cemiplimab, and dostarlimab) and three anti-PD-L1 antibodies (atezolizumab, durvalumab, and avelumab) for the treatment of nearly all types of cancers [1,2,3,4]. Despite these advances in cancer treatment, only a small subset of cancer patients actually benefits from anti-PD-(L)1 immunotherapy either due to tumor cells not responding to the therapy (primary resistance) or due to tumor cells developing resistance to the therapy (acquired resistance) [1,5,6,7,8,9]. Even worse, treatment with anti-PD-(L)1 immunotherapy can lead to an acceleration of tumor growth, defined as Hyperprogressive Disease [10,11,12]. Moreover, many patients terminate treatment because of therapy-induced severe toxicities that are associated with side effects caused by immune activation, referred to as immune related adverse events (irAEs) [13,14]. Therefore, identification of biomarkers to stratify patients who are most likely to benefit from the anti-PD-(L)1 immunotherapy is of critical importance.
Of the three FDA-approved predictive biomarker tests for anti-PD-(L)1 immunotherapy, PD-L1 assessed in tumor tissues by immunohistochemistry (IHC) is the most commonly used biomarker for patient stratification [15,16]. The biological rationale for this is based on the mechanism of action of anti-PD-(L)1 immunotherapy, in which generation of the effective anti-tumor specific T cell immune response requires two signals: (1) T cell priming provided by the T cell receptor (TCR) recognizing and binding to antigen peptide in complex with human leukocyte antigen (HLA) molecules. Along this line, non-germline mutations in TMB-H (high tumor mutation burden) and/or MSI-H (high microsatellite instability) tumors generate tumor-specific neoantigens. This provides a primary signal by forming neo-antigen peptide/HLA complexes that are recognized by TCR, resulting in initiating activation of tumor-specific T cells [17,18,19]. (2) Full T cell activation provided by engagement of co-stimulatory receptors expressed on T cells and its ligands expressed on antigen presenting cells (APCs), such as co-stimulatory receptor CD28, and its ligands CD80 (B7-1) and CD86 (B7-2). PD-L1 expressed on tumor cells and/or tumor stromal cells impedes this second signal by engagement of PD-1 expressed on T cells, resulting in dysfunction of anti-cancer T cell responses [20,21,22,23].
PD-L1 expression on tumor tissues has clearly shown the predictive value in many types of cancers, as patient responses to anti-PD-(L)1 immunotherapy are linearly associated with increased levels of PD-L1 expression in many types of cancers [24,25,26]. However, positive PD-L1 expression can only partially predict which patients benefit from therapy, as a subset of patients whose tumors lack expression of PD-L1 has also been shown to respond positively to anti-PD-(L)1 immunotherapy. Moreover, most cancer patients do not achieve a durable response regardless of levels of PD-L1 expression [15,27]. The inability of tumor biopsy to capture the complexity of intra-tumor heterogeneity and heterogenous phenotypes in tumor tissues (spatial limitation) is a major attribute of the imperfection of PD-L1 use as a biomarker [28,29,30]. Additional challenges to this approach include difficulties in acquiring tumor biopsies, such as early stage pancreatic cancer tissues and their unsuitability for longitudinal monitoring of the dynamic changes of tumor cells and other stromal cells within the tumor microenvironment (temporal limitation) [27,31]. Lastly, it is challenging to harmonize the assays to assess PD-L1 expression in tumor tissues, including how to select an appropriate PD-L1 assay platform and how to score the PD-L1 expression consistently and accurately on tumor cells, immune cells, or both [32,33,34]. An alternative approach, using liquid biopsies to analyze PD-L1 expression on cytological samples has shown promise, which has the potential to overcome many of the challenges observed when using solid tumor biopsies [35,36].
While enormous efforts have been made for improving the quality and accuracy of PD-L1 assessments using tumor biopsy-based approaches [37], recent advances in liquid biopsy might not only overcome the many hurdles facing the field of identification of biomarkers in tumor biopsies, but also provide opportunities to discover novel biomarkers or to improve the accuracy of currently approved biomarkers. The rationale for using blood-based biopsies for identifying biomarkers is that nearly all components in the tumor microenvironment can be found in blood. These components include genomic material, proteins, metabolites, and extracellular vesicles, as well as various cell types including circulating tumor cells (CTCs) and immune cells trafficking between tumors to the blood stream and lymphoid organs [38]. In comparison with tumor biopsy, a blood-based biopsy approach has the following advantages: (1) the convenience of sampling can be used to longitudinally monitor the dynamic biological changes within the tumor microenvironment; (2) samples in the blood contain components derived from both primary and metastatic tumor tissues; (3) ease of development and validation of many assays for testing multiple blood components; and (4) systemic anti-tumor T cell responses have recently emerged as a promising potential biomarker, which can only be tested using peripheral blood mononuclear cells [39].
In this review, we provide an overview of recent advances in understanding the complexity of regulation and function of PD-L1 expression. We discuss the rationale for using different components of blood to assess PD-L1 as a biomarker with a focus on the mechanistic and technical challenges involved. Lastly, we discuss the key challenges and prospects in the development and validation of blood-based PD-L1 assessments for anti-PD-(L)1 immunotherapy based on our evolving understanding of the mechanisms of action underlying this therapy.

2. Regulation and Function of PD-L1 Expression

PD-L1 (B7-H1, CD274) is a type 1 transmembrane glycoprotein encoded by the CD274 gene. PD-L1 is highly expressed on tumor cells, as well as on hematopoietic cells, including macrophages, dendritic cells, mast cells, neutrophils, myeloid-derived suppressive cells (MDSCs), platelets, and T and B lymphocytes, and on non-hematopoietic cells, such as epithelial cells, endothelial cells, mesenchymal stem cells, placental trophoblasts, and many others [15]. Expression of PD-L1 is regulated at the transcriptional, post-transcriptional, and post-translational levels [40,41,42,43]. At the transcriptional level, PD-L1 expression is modulated by JAK/STAT1, NF-kappa B, and other transcriptional factors. Additionally, glycosylation and acetylation of PD-L1 are two important post-translational regulators of PD-L1 expression. For example, acetylation of PD-L1 impacts the translocation of PD-L1 from the cytoplasm to the nucleus, whereas glycosylation of PD-L1 can enhance its ability to interact with PD-1 to increase anti-cancer T cell immune responses [44,45]. Expression of PD-L1 is also impacted by protein degradation pathways such as autophagic degradation and ubiquitination. PD-L1 degradation pathways not only affect the efficacy of anti-PD-(L)1 immunotherapy, but also affect its pharmacokinetics and pharmacodynamics—parameters that are critical for the timing of sampling of PD-L1 for biomarker assessment [46,47,48]. The regulation of PD-L1 expression differs significantly between different cancer types, as well as between tumor cells and non-tumor cells due to variation of aberrations of activation of many signaling pathways such as RAS/RAF/ERK, PI3K/AKT, and JAK/STAT3, among others [41]. Of note, gene amplification, gene translocation, copy-number gains, or copy-number loss also occur in tumor cells [47,49]. PD-L1 copy number changes are associated with PD-L1 expression in several types of cancers such as NSCLCs, urothelial carcinoma, and breast carcinoma, among others. However, PD-L1 gene amplification is not associated with PD-L1 expression when tumor tissues are assessed by IHC in a subset of Hodgkin lymphoma patients [50,51,52,53].
The major function of PD-L1 is suppression of T cell activation via extrinsic binding to its cognate receptor PD-1 expressed on T cells and subsequent downregulation of T cell-mediated anti-tumor responses in general, thereby resulting in a concomitant inhibition of T cell-mediated anti-tumor responses. Recently, intrinsic PD-L1 signaling has been shown to also play a critical role in promoting tumor progression, metastasis, immune evasion, and inducing responsiveness to anti-PD-(L)1 immunotherapy via a PD-1 dependent or independent means [54,55,56,57]. The cell-intrinsic PD-L1 signaling can be modulated by: (1) its post-translational modifications such as acetylation or glycosylation, or by forming cis-heterodimers (i.e., the ligand binding to receptors expressed on the same cell), such as PD-L1/PD-1 or PD-L1/CD80 heterodimers (CD80 being a shared receptor for CTLA4 and CD28); (2) PD-L1 signaling in cancer cells triggered by engagement of PD-1; and (3) PD-L1 binding to co-receptor integrins. From the perspective of biomarker identification, the cell-intrinsic PD-L1 signaling dictates what assays should be used for testing PD-L1 expression. For example, anti-glycosylated PD-L1 specific antibody needs to be used for testing expression of glycosylated PD-L1 on cells, whereas an intracellular staining protocol is needed to assess acetylated PD-L1 located in the cell’s nucleus.
PD-L1 can be cleaved by cell surface proteases such as A Disintegrin. Additionally, Metalloproteinase 10 and 17 (ADAM10 and ADAM17) and the cleaved form of PD-L1 (cPD-L1) can be detected in the serum or plasma of patients with NSCLC, melanoma, and many other cancers [58,59,60]. Several PD-L1 splice variants (sPD-L1) can also be detected in plasma of patients with NSCLC and other cancer types [61,62,63,64]. In addition, the expression levels of PD-L1 in serum or plasma or other components detected in blood include circulating tumor cells (CTCs), exosomes, and various immune cells that have been assessed and proposed to be a predictive biomarker for melanoma, NSCLC, gastrointestinal cancer, breast cancer, and many others [27,65,66,67,68,69,70]. In the following report, we discuss the potential use of PD-L1 expression in different blood components as a predictive biomarker for anti-PD-(L)1 immunotherapy with a focus on addressing the mechanistic and technical challenges that are based on our understanding of the heterogeneity and complexity of the PD-L1 regulation in cancer cells and immune cells.

3. PD-L1 on Circulating Tumor Cells

Circulating tumor cells (CTCs) are rare cancer cells found in the blood that are derived from solid tumors. The rationale for using PD-L1 positive CTCs (PD-L1+CTCs) as a potential biomarker include: (1) PD-L1 expressed on CTCs plays an important role in tumor progression and metastasis, as well as resistance of anti-PD-(L)1 immunotherapy [71,72]. (2) CTCs derived from primary tumors or metastatic lesions can be detected in blood, providing the opportunity that assessment of the levels of PD-L1 expression in CTCs might recapitulate the PD-L1 expression pattens in tumor tissues [15]. This is important since that it has been shown that the levels of PD-L1 expression are not equal between primary tumors and metastatic tumors in many patients [73]. PD-L1 expression in metastatic tumors, but not in primary tumors, is associated with poor prognosis of melanoma patients [74]. (3) The number of CTCs has been shown to be a prognostic biomarker for NSCLC and many other cancers and is also associated with the tumor responses to anti-PD-(L)1 immunotherapy [75,76]. Finally, (4) many assays that are currently available to assess the PD-L1+CTCs are imperfect and are facing many technical challenges (discussed below). Therefore, assessment of the number of PD-L1+CTCs as a biomarker is currently being explored in NSCLC, melanoma, and many other cancers [70,77,78,79].
The association between the number of baseline PD-L1+CTCs and the efficacy of anti-PD-(L)1 immunotherapy is inconclusive and conflicting. Several studies demonstrate that NSCLC patients with high baseline PD-L1+CTCs respond worse to treatment with nivolumab or pembrolizumab [77,79,80,81,82,83]. Similar findings are also reported in melanoma and other types of cancers. However, other studies indicate that baseline numbers of PD-L1+CTCs are not associated with patient responsiveness to nivolumab, including NSCLC patients. In addition, some studies demonstrate that the number of PD-L1+CTCs is correlated with PD-L1 expression in primary tissues, whereases no correlation was found in other studies [77,80,82,84,85]. Moreover, the detection rates of PD-L1+CTCs and its cut-off value in many types of cancers including lung cancer vary dramatically, ranging from less than 30% to 90% across different studies [76,80,84,85,86,87]. The number of PD-L1+CTCs from different patients within the same study also vary dramatically, ranging from one cell to hundreds of CTCs in blood samples from prostate cancer patients, or from one cell to 20 in patients with colorectal cancer [84,85,88]. The median percentage of PD-L1 positive cells from different tumors also varies significantly [89].
The inconsistency and heterogeneity of PD-L1+CTCs highlight the challenge of reconciling the results of these clinical studies. Among several factors that may contribute to the inconsistency, some of them are listed here. (1) Different CTC isolation platforms were used in these studies. It has been shown that the CTC isolation yield differs significantly when different platforms are used [90,91]. Although the CellSearch system (an affinity-based assay for enrichment of EpCAM positive tumor cells) is considered the gold standard CTC detection platform [92], many challenges remain in isolating the rare and highly heterogeneous CTC population [93,94,95]. For example, the CellSearch system cannot be used for isolating EpCAM negative CTCs due to epithelial-to-mesenchymal transition [96]. The procedures and challenges of isolating CTC have been comprehensively reviewed elsewhere [90,95,97,98]. (2) The described studies are retrospective analyses with low statistical power due to the limited sample size. (3) The results from tumor biopsies demonstrate that PD-L1 is a not a universal biomarker for different lines of therapy. For example, PD-L1 is a predictive biomarker for first-line atezolizumab treatment for NSCLC patients, but the anti-cancer efficacy is not associated with PD-L1 expression for second-line atezolizumab treatment [15,99,100]. However, many studies combined mixed lines of treatments, including first-line to multiple line treatment, making it difficult to interrogate data from these studies. (4) The predictive effect of PD-L1 is significantly different among different drugs and cancer types. For example, PD-L1 is a predictive biomarker for first-line and second-line pembrolizumab treatment for NSCLC patients, but not for nivolumab treatment in the same setting [15]. Several studies mentioned above included patients treated with different anti-PD-1 therapeutic antibodies, making it difficult to interrogate data from these studies as well.
Advances in understanding the biology and regulation of PD-L1 might provide additional opportunities to explore the potential use of PD-L1+CTCs as a biomarker. These options include: (1) testing PD-L1+CTCs from different sources such as obtaining tissues from primary tumors, draining lymph nodes, or tumor metastases. It has already been shown that PD-L1 expression can vary significantly between these different cell sources [97,101,102]. The functions and heterogeneity of PD-L1+CTCs suggest that different sources of PD-L1+CTCs may be used as a biomarker. (2) Testing post-translational modifications of PD-L1, such as acetylation or glycosylation of PD-L1, or its nuclear localization. It has been shown that nuclear expression of PD-L1 may be associated with a poorer prognosis in late stage colorectal or prostate cancer, whereas the nuclear expression is not associated with CTCs. In addition, it has been shown that levels of deglycosylated PD-L1 may have a better predictive value for anti-PD-(L)1 immunotherapy than that of glycosylated PD-L1 when tumor biopsy samples were used [103,104,105,106]. (3) CTCs can form CTC clusters. These clusters have shown more metastatic potential than single CTCs in breast cancer and other types of cancers and play a role in cell survival and immune evasion [107,108]. Higher levels of CTC clusters are associated with a worse prognosis in breast cancer patients [109]. In addition, CTCs have also been shown to form clusters with a subset of neutrophils, resulting in the promotion of cancer cell metastasis. Overall, increased levels of CTC–neutrophil clusters are associated with a poorer prognosis in breast cancer patients [110], suggesting that the PD-L1 positive CTCs or CTCs/immune cell clusters might also have a potential to be a biomarker for anti-PD-(L)1 immunotherapy [111,112]. (4) Combining the use of PD-L1+CTCs with other biomarkers of immune checkpoint receptors. The formation of cis-PD-L1/CD80 heterodimers significantly impacts the efficacy of anti-PD-(L)1 immunotherapy, suggesting that co-expression of CD80 and PD-L1 in CTCs might be a useful biomarker [113]. However, it is unlikely that combining PD-L1+CTCs with the approved biomarkers TMB-H or MSI-H would have a predictive value, as multiple studies, based on tumor biopsy results, have demonstrated that PD-L1 is an independent biomarker from TMB-H or MSI-H [114,115,116,117]. Finally, (5) PD-L1 gene amplification is associated with patient responsiveness to anti-PD-(L)1 immunotherapy [53,118], and vice versa [119]. Therefore, the level of PD-L1 gene expression in CTCs could be a useful biomarker and is worth pursuing. The rationale and clinical and technical challenges for using PD-L1+CTCs are summarized in Table 1 below.
As presented in Table 1, advances in development of a reproducible, accurate, and sensitive method to enrich CTCs regardless of cell sources will facilitate overcoming many of the hurdles that currently exist in the use of PD-L1+CTCs, either alone or in combination with other existing biomarkers, to predict treatment responsiveness [91,97,98,120,121,122,123,124,125,126].

4. PD-L1 on Circulating Immune Cells

Numerous efforts have been made to explore whether PD-L1 expression on peripheral immune cells can be used as a biomarker for anti-PD-(L)1 immunotherapy. Several lines of evidence support PD-L1 expressed on circulating immune cells as a biomarker for anti-PD-(L)1 immunotherapy. First, nearly all types of immune cells participate in tumor development, progression, metastasis, and resistance to anti-cancer therapies including anti-PD-(L)1 immunotherapy [39,127,128,129,130,131,132,133]. Second, the levels of PD-L1 positive immune cells alone in tumor tissues have prognostic value for many cancer types such as NSCLS, melanoma, renal carcinoma, and many others [134,135]. Moreover, the levels of PD-L1 expression on tumor-infiltrating immune cells or in combination with PD-L1 expression on tumor cells as assessed by IHC are associated with efficacy of atezolizumab or pembrolizumab treatment [15]. Third, the increased microvascular permeability in the tumor tissue promotes the immune cells trafficking to and from the blood stream, suggesting that PD-L1-positive immune cells from tumor tissues can be detected in blood [136,137,138]. Supporting this, it has been shown that T cells expanded in the tumor microenvironment are present in peripheral blood mononuclear cells (PBMCs) [139].
The current paradigm regarding the mechanisms underlying anti-PD-(L)1 immunotherapy is that the effective anti-tumor T cell responses are dependent on activation of the dysfunctional tumor-infiltrating CD8+ T cells in the tumor microenvironment. Accumulating evidence demonstrates that systemic immunity (defined here as activation of immune responses independent of activation of tumor-infiltrating immune cells) has emerged to be critical for immune-mediated tumor eradication [9,39]. Spitzer et al. demonstrate in a mouse tumor model that activation of T cells in the draining lymph node, bone marrow, and blood contributes to tumor eradication that is independent of the anti-tumor immune response in the tumor microenvironment [140]. Supporting this notion, analyses by single-cell RNA sequencing for T cell receptors in tumor tissues demonstrate that T cells in the tumors are replenished by T cells activated outside of the tumor tissues [139]. Lastly, activated T cells or other immune cells can be found in many organs, including skin, liver, and heart, among others, in patients who develop irAE. Although the origins of these immune cells are not clear, it is possible that activated immune cells from these organs might also be detected in the peripheral blood [141,142,143]. Nonetheless, testing for PD-L1 expression on immune cells in peripheral blood is an attractive approach, as PBMCs might contain both tumor-infiltrating immune cells as well as immune cells activated in the periphery by anti-PD-(L)1 immunotherapy [144,145,146].

4.1. PD-L1 Expression on Myeloid Cells

Myeloid cells including neutrophils, monocytes/macrophages, myeloid-derived suppressor cells (MDSCs), and dendritic cells are important modulators of anti-cancer T cell responses [147,148]. The outcomes of these cell-mediated anti-tumor responses are context dependent. For example, macrophages are a type of antigen-presenting cell, but also contribute to a pro-tumor microenvironment. Expression of PD-L1 in these cells in tumor biopsies is associated with the outcomes of patients, as well as patient responses to anti-PD-(L)1 immunotherapy. For biomarker identification, infiltration of myeloid cells has been proposed as a biomarker for anti-PD-(L)1 immunotherapy [149]. We will discuss the opportunities and challenges of assessing expression of PD-L1 in each of these cells as a biomarker for anti-PD-(L)1 immunotherapy.

4.1.1. Neutrophils

Neutrophils are the most abundant of the white blood cells and are a major part of the innate immune response against infections. The phenotypes and functions of tumor-associated neutrophils (TANs) are highly heterogeneous. TANs display either anti-tumor N1 or pro-tumor N2 phenotype or mixed N1 and N2 phenotypes. These phenotypes are dictated by interaction with tumor cells and other stromal cells in the tumor microenvironment [150,151,152,153,154]. As neutrophil and lymphocyte counts are routinely conducted in clinical settings, the neutrophil-to-lymphocyte ratio (NLR) is one of most studied prognostic biomarkers for cancer progression and anti-cancer therapies including anti-PD-(L)1 immunotherapy [155,156]. The rationale for using NLR is based on multiple studies that demonstrate that the numbers of neutrophils or lymphocytes are associated with poorer, or better outcomes for cancer patients, resulting in amplifying the predictive effects. While many studies demonstrate that cancer patients with higher NLR have a poor prognosis in multiple tumor types or respond poorly to anti-PD-(L)1 immunotherapy, other studies have shown the opposite predictive effects [150,157,158]. To improve its predictive power, combining NLR with other biomarkers has also been explored. A retrospective study by Valero et al. demonstrated that a higher NLR is associated with significantly poorer efficacy of anti-PD-(L)1 immunotherapy in 16 different types of cancers. Combination of NLR with TMB has better predictive effects than NLR alone, and patients that are NLR low/TMB high respond better to anti-PD-(L)1 immunotherapy than patients that are NLR high/TMB high. [159]. Combining NLR with other factors appears to be a better predictive biomarker than NLR alone for NSCLCs patients treated with anti-PD-(L)1 immunotherapy [160,161,162,163,164,165]. In line with the baseline assessment, patients with high NLR after anti-PD-(L)1 immunotherapy respond poorly to anti-PD-(L)1 immunotherapy [166,167].
As Mentioned, combining NLR with other factors such as platelet-to lymphocyte ratio appears to improve the predictive abilities [168,169,170]. Conversely, the combination of NLR with PD-L1 tumor proportion score as assessed by IHC or soluble PD-L1 in the blood is inversely associated with the efficacy of anti-PD-(L)1 immunotherapy [171]. PD-L1 positive neutrophils, either alone or in combination with other biomarkers might be worthy of further investigation.

4.1.2. Monocytes and Macrophages

Macrophages are among the most abundant of immune cells in the tumor microenvironment [172,173]. Tumor-associated macrophages (TAMs) are highly heterogeneous, having shown a mixed anti-tumor M1 phenotype and pro-tumor M2 phenotype. TAMs are differentiated from monocytes and produce numerous growth factors, cytokines, and chemokines to modulate tumor initiation, progression, metastasis, modulation of anti-cancer immune responses, and responses to anti-cancer therapies including PD-(L)1 immunotherapy [133,173]. Several lines of evidence suggest that PD-L1-positive monocytes/macrophages are a potential biomarker for anti-PD-(L)1 immunotherapy. First, multiple studies have shown that most TAMs are M2-like macrophages and that increased numbers of TAMs correlate with a poor prognostic outcome [135,174]. Second, some studies demonstrate that PD-L1 is more highly expressed on immune cells than on tumor cells, and in some cases, PD-L1+TAMs are among the most predominant immune cells in the tumor microenvironment [175,176]. Third, while most studies demonstrate that PD-L1+ TAMs are immunosuppressive and show pro-tumor phenotypes, other studies suggest that baseline PD-L1+ TAMs do not contribute to suppression of anti-tumor T cell responses and in some cases are associated with better prognosis [177,178,179,180]. Similarly, some studies have demonstrated that the numbers of PD-L1+ TAMs or PD-L1+ monocytes are associated with tumor progression and inversely associated with patient prognosis in multiple cancer types [181,182,183], whereas others studies have demonstrated that baseline numbers of PD-L1+ TAMs do not have predictive value for melanoma patients treated with anti-PD-1 immunotherapy [184]. However, in melanoma, the patients who responded better to anti-PD-1 immunotherapy had increased numbers of PD-L1+ TAMs [184,185]. Finally, peri-tumor monocytes/macrophages, which could be a potential source of peripheral monocytes, have also been shown to have some prognostic value. Similar to PD-L1+ TAMs, it has been shown that peritumor PD-L1+ monocytes/macrophages are also immunosuppressive and show a pro-tumor phenotype [181,186].
The predictive value of circulating PD-L1+ monocytes/macrophages for anti-PD-(L)1 remains less studied and inconclusive. While it has been reported that metastatic melanoma patients with CD14+CD16HLA-DRhi monocytes in the peripheral blood prior to treatment respond poorly to anti-PD-1 immune checkpoint therapy [9,187,188], other studies demonstrated that the frequency of PD-L1+ CD14+ monocytes in the peripheral blood is inversely associated with patient responses to anti-PD-1 immunotherapy [189,190]. Moreover, one study has shown that PD-L1 expression on non-classical (CD14dimCD16+) and intermediate (CD14+CD16+) monocytes is significantly increased in patients and is associated with efficacy of melanoma patients treated with anti-PD-1 immunotherapy [191].

4.1.3. Myeloid-Derived Suppressor Cells

Myeloid-derived suppressor cells (MDSCs) are highly immunosuppressive cells derived from immature myeloid progenitor cells [192]. Based on their origin, function, and surface markers, MDSCs are subdivided into two major subsets: polymorphonuclear-MDSCs (PMN-MDSCs) and monocytic MDSCs (M-MDSCs). Cytokines involved in myeloid cell development and differentiation such as GM-CSF, G-CSF, M-CSF, IL-6, and others are major modulators for differentiating myeloid progenitor cells into PMN-MDSCs and M-MDSCs. MDSCs exert their immunosuppressive functions by secreting immunosuppressive factors, including adenosine, IL-10, and TGF-β, as well as increasing activation of regulator T cells. Depletion of MDSCs or targeting MDSCs with pharmaceutical intervention increases the efficacy of anti-PD-(L)1 immunotherapy [192,193,194].
PD-L1 is also expressed on PMN-MDSC and M-MDSCs and contributes to MDSC-mediated immunosuppressive effects [195,196,197]. The predictive roles of PD-L1+ MDSCs as a biomarker for anti-PD-(L)1 immunotherapy is less studied, although it has been shown that cancer patients with high baseline levels of circulating PMN-MDSC or M-MDSCs respond better to anti-PD-1 immunotherapy treatment [191,198]. It is unlikely that circulating PD-L1+MDSCs alone can be a biomarker for anti-PD-(L)1 immunotherapy, as MDSCs produce numerous immunosuppressive factors and cytokines. Thus, PD-L1 expressed on MDSCs may only be partially contributing to MDSC-mediated immunosuppressive effects (i.e., TIM3 ligand Galectin-9 expressed on MDSCs also contributes to MDSC-mediated immunosuppression [199]).

4.1.4. Dendritic Cells

Dendritic cells (DCs) are the most potent of the antigen-presenting cells and play essential roles in anti-tumor immunity via priming and maintaining effective T-cell-mediated anti-tumor immunity, as well as other innate and adaptive anti-tumor effects [134,200,201,202,203,204]. All subtypes of DCs can be detected in the peripheral blood [190,203]. Based on their morphology, phenotype, and functions, DCs are subdivided into three major subtypes: conventional type 1 dendritic cells (cDC1s), conventional type 2 dendritic cells (cDC2s), and plasmacytoid dendritic cell (pDC) [205,206,207]. Tumor-associated DCs are generally not fully functional and thus cannot truly activate anti-tumor T cell immunity [202,203,204,208,209,210]. Several lines of evidence suggest that PD-L1+ DCs might be a potential biomarker for anti-PD-(L)1 immunotherapy. First, the numbers and activation level of tumor-associated DCs have been shown to be a good prognostic indicator in several cancer types [211,212,213,214]. Second, studies from several groups clearly demonstrate that PD-L1 expressed on dendritic cells contributes to inhibition of effective anti-tumor T cell responses. Targeting PD-L1 expressed on DCs using different approaches can, in some cases, stimulate the anti-tumor T cell response to eradicate tumor cells [57,214,215,216,217]. Additionally, PD-L1 intrinsic signaling is involved in DC migration, resulting in a deficiency in T cell priming [218]. Fourth, it has been shown that PD-L1+ DCs are associated with a favorable patient outcome and thus may be a good biomarker for stage III colon cancer [219]. Overall, PD-L1 expression on DCs, rather than on tumor cells, appears to be critical for anti-PD-(L)1 treatment-induced anti-tumor immunity [220]. In contrast, another study demonstrated that PD-L1 expressed on cDCs in tumor-draining lymph nodes, but not in the tumor microenvironment, is associated with a poor prognosis in melanoma [221].
The predictive roles of circulating PD-L1+ cDCs have also not been fully investigated. Liu et al. reported that ovarian cancer and melanoma patients with high expression of PD-L1 on dendritic cells respond better to anti-PD-(L)1 immunotherapy [222]. However, another study reported that patients with high PD-L1 blood DC subtypes respond poorly to PD1 inhibitor therapy [190].

4.2. PD-L1 Expression on Lymphocytes

Tumor-infiltrating lymphocytes (TILs) are one of the most studied biomarkers for anti-PD-(L)1 immunotherapy due to the fact that all individual types of lymphocytes (including CD4+ T cells, CD8+ T cells, B cells, NK cells, and others) have been shown to be critical for effective anti-tumor immunity and that TILs are routinely identified in tumor tissues simply stained with hematoxylin and eosin (H&E) [9,223,224,225,226,227,228,229]. Patients with higher numbers of TILs are generally associated with favorable outcomes in many types of cancers, with a few exceptions, and respond better to anti-PD-(L)1 immunotherapy [229,230]. Of note, the combination of PD-L1 expression in tumor tissues with TILs has also shown a prognostic value for anti-PD-(L)1 immunotherapy [223,224].
Nearly all types of TILs were evaluated as a biomarker for anti-PD-(L)1 immunotherapy. Briefly, cancer patients with high numbers of baseline tumor-infiltrating total CD4+ T cells, Th1 CD4+ T cells, CD8+ T cells, the ratio of CD8+ T cells/CD4+ T cells, T follicular helper-T cells (Tfh), Th1, Th9, and Th17 CD4+ T cells respond better to anti-PD-1 immunotherapy, whereas an inverse predictive value was observed in patients with high levels of infiltrating regulatory T cells or Th2 CD4+ T cells [226,231]. Further analyzing the association of activation state between TILs with the patient responsiveness to anti-PD-(L)1 immunotherapy indicated that: (1) a higher ratio of tumor-infiltrating Tfh/exhausted CD8+ T cells is associated with favorable outcomes for NSCLC patients treated with anti-PD-1 immunotherapy [232]; (2) melanoma patients with a higher percentage of tumor-infiltrating progenitor exhausted CD8+ T cells (express intermediate of PD-1, CXCR5), but not terminally exhausted CD8+ T cells (express high level of PD-1 and other co-inhibitory receptors such as TIM3), respond better to anti-PD-1 immunotherapy [233]; (3) a combination of signatures of T cell dysfunction and the numbers of infiltrating CD8+ T cells has better predictive value than individual PD-L1 levels or TMB for evaluating the efficacy of anti-PD-1 or anti-CTLA-4 immunotherapy [234]; and (4) cancer patients with high levels of tissue-resident memory CD8+ T cells (CD8+ TRMs) are associated with favorable outcomes and respond better to anti-PD-(L)1 immunotherapy [235].
The predictive values of circulating T cells, in particular circulating PD-L1+ T cells, have not been fully investigated, and the results from different studies have yielded different conclusions [236,237,238]. Furthermore, the frequency of circulating PD-L1+ T cells (less than 1% of PBMCs for CD3) is significantly lower than for myeloid cells, which poses the technical challenges of developing accurate and robust assays capable of assessing the frequency of circulating PD-L1+ T cells. However, it is still worth exploring the predictive value of PD-L1 expression on T cells due to the following: (1) circulating T cells can recapitulate many phenotypes and functional characteristic of TILs, such as matched TCRαβ repertoire, or the gene signatures of effector functions [239]. Moreover, characterization of T cell signatures from patients treated with atezolizumab suggested that activated tumor-infiltrating CD8+ T cells may be derived from T cells expanded in the periphery [139]. (2) A number of different types of T cells may be detected in peripheral blood including CD8+ TRMs [235], Treg cells [240], and mucosal-associated invariant T cells [241] among others. (3) The frequencies of baseline individual circulating T cell types are associated with responsiveness to anti-PL-(L)1 immunotherapy [241,242]. (4) PD-L1 expressed on T cells has been shown to be a highly immunosuppressive phenotype and can significantly inhibit anti-cancer immunity [243]. Indeed, it has been reported that expression of PD-L1 on circulating T cells is higher in melanoma patients compared to healthy donors, and patients with lower baseline levels of circulating PD-L1+ CD4+ and CD8+ T cells respond better to ipilimumab [244].
In addition to demonstrating the role of baseline levels of circulating PD-L1+ T cells in ICI-mediated cancer treatments, assessing the levels of circulating PD-L1+ T cells after patients are treated with anti-PD-(L)1 immunotherapy has emerged as a new approach to investigate the underlying mechanism and biomarker potential of these molecular markers. The rationale for using on-treatment blood samples is based on the following: (1) expression of PD-L1 is upregulated by IFN-γ, and the IFN-γ signature is associated with favorable outcomes for patients treated with anti-PD-(L)1 immunotherapy [245,246,247]; (2) treatment with anti-PD-(L)1 immunotherapy significantly increases the numbers of circulating CD4+ and CD8 T+ cells [248], as well as the frequency of PD-L1+ T cells. However, in a dose-escalation phase I trial, it was observed that treatment with avelumab (the only IgG1 mAb; other anti-PD-(L)1 mAbs are all IgG4) did not impact the frequencies of circulating PD-L1+CD4+ and CD8+ T cells in 12 different cancer types. This result needs to be carefully interpreted because of the small sample size (only one patient was evaluated in lung, prostate, ovarian, and other cancers), and the dose of avelumab was not optimized. Given the potential use of the activation state of a patient’s T cells after treatment as a biomarker, we propose that an in vitro PBMC-based assay can be potentially used as a biomarker for anti-PD-(L)1 immunotherapy. For this model, we evaluated T cell functions after nivolumab treatment in the presence of a suboptimal anti-CD3 mAb (non-specific stimulation of TCR signaling). We found that T cell responses to nivolumab vary significantly among different donors. The major rationale for using our model is that T cell responses to nivolumab are similar to those observed in patients treated with anti-PD-(L)1 immunotherapy such as elevated cytokine production, increased proliferative T cells, and increased expression of PD-L1 and many co-inhibitory receptors ([249,250,251,252] and unpublished data), and that activation of T cells by anti-PD-(L)1 immunotherapy might not be tumor antigen-dependent in some cancer patients. Supporting this notion, it was found that PD-L1 is an independent biomarker from TMB in most types of cancers [115]. Renal carcinoma, for example, has a low TMB, but responds well to anti-PD-(L)1 immunotherapy [253].
In addition to T cells, accumulating evidence demonstrates that other types of lymphocytes, including B cells and NK cells as well as tertiary lymphoid structures (TLS, a lymph node-like structure found in the tumor), play critical roles in anti-cancer immunity and are associated with the efficacy of anti-PD-(L)1 immunotherapy. It has been demonstrated that tumor-associated B cells, plasma cells, or TLS are presented in the tumor microenvironment and are associated with improved survival and immunotherapy responsiveness in many types of cancers independent of TILs and PD-L1 expression in tumor tissues [254,255,256,257,258]. Additionally, although the numbers of tumor-infiltrating NK cells (TINKs) is less than T cells and B cells, cancer patients with higher baseline numbers of TINK cells generally have more favorable outcomes and respond better to anti-PD-(L)1 immunotherapy [259,260]. Lastly, different B cell subtypes such as B1B cells, regulatory B cells, also play a role in modulating anti-cancer immunity [261]. Collectively, it is possible that the frequency of circulating NK cells and B cells could also be used for assessing their potential as a predictive biomarker.
The combination of circulating PD-L1+ T cells with myeloid cells or tumor cells as a biomarker has also been explored [262]. However, it is challenging to find the right combinations as more available parameters become available. For example, analysis of TILs using mass cytometry has identified 22 different T cell subsets and 17 macrophage subsets in renal carcinoma [263,264]. In another study, a total of 123 immune cell subsets (including circulating PD-L1+ immune cells) were identified [265]. Recent advances in artificial intelligence for diagnostic and machine learning techniques will surely provide the opportunity to dissect the complexity and heterogeneity of TILs, as well as dynamic temporal changes of PD-L1 expression on immune cells for biomarker identification [266,267,268,269,270,271]. Expression of PD-L1 on circulating immune cells is unlikely to be useful as a stand-alone biomarker as they are not consistent with those on tumor infiltration immune cells (i.e., TLS only can be found in tumor tissues), and one cannot spatially differentiate tumor-infiltrating PD-L1+ immune cells from tumor cells [272,273]. The rationale and clinical and technical challenges in the use of PD-L1-positive immune cells as a biomarker are summarized in Table 2 below.

5. Circulating Exosomal PD-L1

Exosomes are a subset of extracellular membrane-bound vesicles of endosomal origin. Exosomes detected in tumor patients can be derived from all cell types in the primary tumor or metastatic tissues including tumor cells, immune cells, and other stromal cells, such as fibroblasts in the tumor microenvironment, as well as normal cells outside of any tumor [274]. Exosomes serve as a messenger between different cells in the tumor microenvironment by transferring cellular cargoes to the recipient cells [275,276]. Tumor-derived exosomes participate in every stage of cancer development and in modulating anti-cancer therapies via functional proteins, genetic materials such as mRNAs, long non-coding RNAs, and DNA fragments, as well as metabolites [275,276,277,278,279,280,281,282,283]. PD-L1 is highly expressed on tumor-derived exosomes and functions similarly to PD-L1 expressed on tumor cells and immune cells. Exosomal PD-L1 (exoPD-L1) is highly immunosuppressive on T cell-mediated anti-tumor immunity in many types of cancers including melanoma, NSCLC, prostate and breast cancers, and many other types of cancers [284,285,286]. Consistent with the functions of exoPD-L1, the levels of exoPD-L1 are inversely associated with clinical outcome, as reported in several studies [287,288].
As gene expression patterns between tumor tissues and exosomes detected in plasma or serum are closely correlated, and circulating exoPD-L1 (cePD-L1) can be detected at the gene and protein level in many types of cancers, much effort has been devoted to exploring the potential use of circulating exoPD-L1 for monitoring the efficacy of anti-PD-(L)1 immunotherapy [68,289]. Several studies suggest that the baseline cePD-L1 levels are inversely correlated with the efficacy of first- or second-line anti-PD-1 or adjuvant anti-PD-(L)1 immunotherapy in metastatic melanoma patients, NSCLC, and others [285]. Another study indicated that there is no association between the levels of cePD-L1 with the clinical benefits of anti-PD-(L)1 immunotherapy. Combinations of exoPD-L1 with other potential biomarkers were also explored for identifying predictive biomarkers. Zhang et al. reported that patients with high exoPD-L1 and low CD28 expressions respond poorly to anti-PD-1 treatment [290]. In addition, since exosomes also express many immune modulators, such as CD9, CD63, CD81, MHC II, and TGF-β, among others, it is conceivable that the combination of exoPD-L1 with these molecules may identify a better predictive biomarker than exoPD-L1 alone [282,291].
Studies on the expression of cePD-L1 and response to anti-PD-(L)1 response remain inconsistent. Several studies have demonstrated that during early stages of anti-PD-1 treatment, there is an increase in the levels of cePD-L1, and increasing magnitudes of cePD-L1 expression are positively associated with better response to the immunotherapy [285,292]. Relevant to this, other studies demonstrated that there is a decrease in the expression levels of PD-L1 mRNA of the plasma-derived exosomes in patients who responded well to anti-PD-1 immunotherapy [66]. Another study suggested that patients with a high increase in cePD-L1 respond poorly to anti-PD-1 immunotherapy, and this is associated with tumor progression [288].
Similar to PD-L1 expressed on tumor cells, there are many technical and mechanistic challenges pertaining to using cePD-L1. The mechanisms and methods for isolating exosomes have been well reviewed elsewhere [293,294], and while significant advances in exosome-based technologies and commercial isolation kits have been developed to increase their sensitivity and accuracy [295,296], there is still a lack of standardized isolation and purification methods for circulating exosomes, especially for characterizing exosomes from different cell origins (tumor cell, immune cells, and fibroblast among others) or from different tumor sites (primary tumor vs. metastatic tumor vs. draining lymph node) [294]. This is important because exoPD-L1 derived from different cell sources functions differently, and this may have different predictive values for anti-PD-(L)1 immunotherapy [280,281]. Additionally, characterization and quantification of cePD-L1 expression by analytical approaches such as flow cytometry-based or ELISA assays among others could be impacted by the method used to isolate the circulating exosomes. Moreover, it has been shown that there is no correlation between exoPD-L1 and PD-L1 assessed by IHC in tumors from NSCLC and melanoma patients. This may be partially due to multiple cell origins of exoPD-L1, and cePD-L1 might be nothing more than an accumulation of exosomal PD-L1 from tumor cells and stromal cells [288,297]. Nonetheless, future work to compare the predictive value between exoPD-L1 and PD-L1 expression in tumors is warranted. The rationale and clinical and technical challenges for using PD-L1+CTCs are summarized in Table 3 below.

6. Circulating Soluble PD-L1

The levels of soluble PD-L1 (sPD-L1) in blood have also been explored as a potential biomarker for anti-PD-(L)1 immunotherapy. sPD-L1 is a pool of PD-L1 that may include the cleaved PD-L1 from the surfaces of cells or PD-L1 variants directly released from cells [61,64,298]. Additionally, since most studies used a low-speed centrifugation and did not use exosome isolation protocols, sPD-L1 detected in these studies should also contain cePD-L1. sPD-L1 may also contain PD-L1 mRNA or DNA released from cells in the tumor microenvironment [292,299,300]. Many studies have shown that high levels of sPD-L1 are generally associated with a poor prognosis in many types of cancers, such as lung cancer, melanoma, and renal cell carcinoma, among others [301,302,303,304].
However, the predictive value of sPD-L1 for anti-PD-(L)1 immunotherapy remains inconclusive and contradictory. While some studies demonstrated that patients with increased sPD-L1 expression respond better to anti-PD-(L)1 immunotherapy, other studies demonstrated that this increased sPD-L1 is not associated with the efficacy of anti-PD-(L)1 immunotherapy. Several studies have demonstrated that high baseline sPD-L1 levels are associated with a worse response for metastatic NSCLC patients treated with anti-PD-(L)1 immunotherapy [305,306]. A systematic review and meta-analysis of NSCLC patients also indicated that high baseline sPD-L1 levels are associated with a poor response to anti-PD-(L)1 immunotherapy in several cancer types [307]. However, other studies demonstrated that metastatic NSCLC patients with high baseline sPD-L1 levels respond better to first- and later-line nivolumab or Pembrolizumab [308]. Moreover, some studies reported that the baseline levels of sPD-L1 do not correlate with efficacy in NSCLC patients treated with anti-PD-(L1) immunotherapy [309,310,311]. Furthermore, treatment with anti-PD-(L)1 also increases levels of sPD-L1 and PD-L1 mRNA in the blood [292].
In summary, most studies have demonstrated that high sPD-L1 levels are associated with a worse response to anti-PD-(L)1 immunotherapy, including NSCLC, in direct contradiction to the notion that high PD-L1 levels in the tumor are a good predictive biomarker for NSCLC. This contradiction may be due to the fact that sPD-L1 contains a mixture of PD-L1 from different cell types within the tumor, different tumor sites, and cells from outside of the tumor as well.

7. Conclusions and Future Perspective

We have discussed the predictive value of blood-based PD-L1 expression at the cellular and molecular levels based on our evolving understanding of the regulation and function of PD-L1. Although promising, there are a few key issues that need to be explored to further understand the potential use of blood-based PD-L1 as a biomarker, one in which only a causal relationship between blood PD-L1 expression and patient responses to anti-PD-(L)1 immunotherapy is currently observed.
In addition to the many challenges discussed above, data from many of the currently available studies need to be cautiously interpreted, which is essential for laying a foundation for leveraging the strength of these studies and for the application of artificial intelligence to analyze resulting data. These challenges include the following: (1) Several published studies mixed data from patients treated with different lines of therapies (first-, second-, and later-line) and/or different treatment regiments (nivolumab, pembrolizumab, ipilimumab, etc.), and/or from cancer patients in different disease stages, which make current studies less informative. (2) While the most significant advantage for blood-based PD-L1 biomarker is its ability to longitudinally monitor the patient’s responses to anti-PD-(L)1 expression, it is important to know that blood drug concentrations have not been analyzed in parallel with testing blood PD-L1 expression in nearly all of these studies. As blood drug levels could significantly affect the accuracy, sensitivity, and specificity of the PD-L1 expression testing results, most of the published studies may not be relevant if drug interference has occurred [312]. (3) There are no reliable validated standard assays for testing blood PD-L1 expression. Moreover, most studies lack various negative and/or positive controls to monitor the reliability and robustness of the assay used for testing blood PD-L1 [313]. (4) The assays used in most of the studies may not be able to detect different PD-L1 variants [53,280], the post translational forms of PD-L1 [54], or PD-L1 expressed in the nuclear or cytoplasm, which may also impact the interpretation of data. Collectively, a successful development of blood-based PD-L1 as a biomarker needs a well-articulated prospective clinical trial [314,315,316,317,318], a reliable testing platform [319], a sound statistical approach [320,321], and a better understanding of the origin of blood PD-L1 and its potential functions.
Given the history and challenges of biomarker identification, as well as the heterogeneity of blood PD-L1 expression and regulation, it is unlikely that a blood-based PD-L1 assessment can be used as a stand-alone biomarker for most patients treated with anti-PD-(L)1 immunotherapy. A combination of blood PD-L1 with other biomarker(s) could improve its predictive value for patient stratification. These potential combinations (in addition to combinations discussed above) could be, but are not limited to: (1) differential blood-based PD-L1 expression, such as expression of PD-L1 on immune cells, cancer cells, and exosomes; (2) other biomarkers representing different mechanisms of action, such as genetic signature [322], immune biomarkers such as serum cytokines, soluble immune regulators (TIM3 and LAG3, etc.), metabolic changes of cancer cells and immune cells [323], epigenetic biomarkers [324], gut microbes, etc. [325,326]; (3) tissue-based biomarkers, including PD-L1 and others such as tertiary lymphoid structures; (4) in combination with various non-invasive imaging technologies, such as positron emission tomography (PET) imaging, etc. [327,328,329,330,331]. Along with liquid biopsy, the explosion in uses of new omic technologies, such as proteomics, genomics, and multi-omics, provides an unprecedented opportunity for biomarker identification. However, it remains challenging to evaluate the reliability of these assays and to interpret the complex array of data obtained [332,333,334,335].
Despite limited success for most proposed biomarkers for anti-PD-(L)1 immunotherapy, we believe that through collaborative efforts by government agencies, industry, and academia [336,337], advances to our deeper understanding of the biology and mechanism of action of anti-PD-(L)1 immunotherapy [127,338,339,340], the application of biomarker-driven clinical trials, efforts to develop robust and sensitive testing technologies, and the application of artificial intelligence for biomarker identification [269,332,341,342] will pave the way for advancing the use of blood-based PD-L1 assessment as part of the armamentarium to deliver the maximal benefits for cancer patients treated with anti-PD-(L)1 immunotherapy.

Author Contributions

T.W.: conception, design, and original draft; D.D. and S.M.B.: critical discussion of the content; G.M.F.: conception, critical intellectual content, and editorial input. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This project was supported in part by the appointment of D.D. to the Research Participation Program at the Center for Drug Evaluation and Research, administered by the Oak Ridge Institute for Science and Education through an interagency agreement between the U.S. Department of Energy and the U.S. Food and Drug Administration. The project is also supported by a grant from the Office of the Chief Scientist, the U.S. Food and Drug Administration, and a grant from the Office of Pharmaceutical Quality Center of Excellence (COE) in Immunology, the U.S. Food and Drug Administration. This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

Disclaimer

The views expressed are those of the authors and do not necessarily represent those of nor imply endorsement from the U.S. Food and Drug Administration or the U.S. government.

References

  1. Morad, G.; Helmink, B.A.; Sharma, P.; Wargo, J.A. Hallmarks of response, resistance, and toxicity to immune checkpoint blockade. Cell 2021, 184, 5309–5337. [Google Scholar] [CrossRef] [PubMed]
  2. Olivier, T.; Haslam, A.; Prasad, V. Anticancer Drugs Approved by the US Food and Drug Administration From 2009 to 2020 According to Their Mechanism of Action. JAMA Netw. Open 2021, 4, e2138793. [Google Scholar] [CrossRef] [PubMed]
  3. Wolchok, J.D.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.-J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Long-Term Outcomes with Nivolumab Plus Ipilimumab or Nivolumab Alone Versus Ipilimumab in Patients with Advanced Melanoma. J. Clin. Oncol. 2022, 40, 127–137. [Google Scholar] [CrossRef] [PubMed]
  4. Korman, A.J.; Garrett-Thomson, S.C.; Lonberg, N. The foundations of immune checkpoint blockade and the ipilimumab approval decennial. Nat. Rev. Drug Discov. 2021. [Google Scholar] [CrossRef] [PubMed]
  5. Kalbasi, A.; Ribas, A. Tumour-intrinsic resistance to immune checkpoint blockade. Nat. Rev. Immunol. 2020, 20, 25–39. [Google Scholar] [CrossRef] [PubMed]
  6. O’Donnell, J.S.; Teng, M.W.L.; Smyth, M.J. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat. Rev. Clin. Oncol. 2019, 16, 151–167. [Google Scholar] [CrossRef]
  7. Pitt, J.M.; Vétizou, M.; Daillère, R.; Roberti, M.P.; Yamazaki, T.; Routy, B.; Lepage, P.; Boneca, I.G.; Chamaillard, M.; Kroemer, G.; et al. Resistance Mechanisms to Immune-Checkpoint Blockade in Cancer: Tumor-Intrinsic and -Extrinsic Factors. Immunity 2016, 44, 1255–1269. [Google Scholar] [CrossRef] [Green Version]
  8. 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]
  9. Bagchi, S.; Yuan, R.; Engleman, E.G. Immune Checkpoint Inhibitors for the Treatment of Cancer: Clinical Impact and Mechanisms of Response and Resistance. Annu. Rev. Pathol. Mech. Dis. 2021, 16, 223–249. [Google Scholar] [CrossRef]
  10. Champiat, S.; Ferrara, R.; Massard, C.; Besse, B.; Marabelle, A.; Soria, J.-C.; Ferté, C. Hyperprogressive disease: Recognizing a novel pattern to improve patient management. Nat. Rev. Clin. Oncol. 2018, 15, 748–762. [Google Scholar] [CrossRef]
  11. Ferrara, R.; Mezquita, L.; Texier, M.; Lahmar, J.; Audigier-Valette, C.; Tessonnier, L.; Mazieres, J.; Zalcman, G.; Brosseau, S.; Le Moulec, S.; et al. Hyperprogressive Disease in Patients with Advanced Non–Small Cell Lung Cancer Treated With PD-1/PD-L1 Inhibitors or With Single-Agent Chemotherapy. JAMA Oncol. 2018, 4, 1543–1552. [Google Scholar] [CrossRef] [PubMed]
  12. Russo, G.L.; Moro, M.; Sommariva, M.; Cancila, V.; Boeri, M.; Centonze, G.; Ferro, S.; Ganzinelli, M.; Gasparini, P.; Huber, V.; et al. Antibody–Fc/FcR Interaction on Macrophages as a Mechanism for Hyperprogressive Disease in Non–small Cell Lung Cancer Subsequent to PD-1/PD-L1 Blockade. Clin. Cancer Res. 2019, 25, 989–999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Davar, D.; Kirkwood, J.M. PD-1 Immune Checkpoint Inhibitors and Immune-Related Adverse Events: Understanding the Upside of the Downside of Checkpoint Blockade. JAMA Oncol. 2019, 5, 942–943. [Google Scholar] [CrossRef] [PubMed]
  14. Martins, F.; Sofiya, L.; Sykiotis, G.P.; Lamine, F.; Maillard, M.; Fraga, M.; Shabafrouz, K.; Ribi, C.; Cairoli, A.; Guex-Crosier, Y.; et al. Adverse effects of immune-checkpoint inhibitors: Epidemiology, management and surveillance. Nat. Rev. Clin. Oncol. 2019, 16, 563–580. [Google Scholar] [CrossRef]
  15. Doroshow, D.B.; Bhalla, S.; Beasley, M.B.; Sholl, L.M.; Kerr, K.M.; Gnjatic, S.; Wistuba, I.I.; Rimm, D.L.; Tsao, M.S.; Hirsch, F.R. PD-L1 as a biomarker of response to immune-checkpoint inhibitors. Nat. Rev. Clin. Oncol. 2021, 18, 345–362. [Google Scholar] [CrossRef]
  16. Sidaway, P. PD-L1 positivity predicts response. Nat. Rev. Clin. Oncol. 2019, 16, 337. [Google Scholar] [CrossRef]
  17. Haen, S.P.; Löffler, M.W.; Rammensee, H.-G.; Brossart, P. Towards new horizons: Characterization, classification and implications of the tumour antigenic repertoire. Nat. Rev. Clin. Oncol. 2020, 17, 595–610. [Google Scholar] [CrossRef]
  18. Jhunjhunwala, S.; Hammer, C.; Delamarre, L. Antigen presentation in cancer: Insights into tumour immunogenicity and immune evasion. Nat. Cancer 2021, 21, 298–312. [Google Scholar] [CrossRef]
  19. Jardim, D.L.; Goodman, A.; de Melo Gagliato, D.; Kurzrock, R. The Challenges of Tumor Mutational Burden as an Immunotherapy Biomarker. Cancer Cell 2021, 39, 154–173. [Google Scholar] [CrossRef]
  20. Topalian, S.L.; Taube, J.M.; Anders, R.A.; Pardoll, D.M. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat. Cancer 2016, 16, 275–287. [Google Scholar] [CrossRef]
  21. Chen, L.; Flies, D.B. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat. Rev. Immunol. 2013, 13, 227–242, Erratum in Nat. Rev. Immunol. 2013, 13, 542. [Google Scholar] [CrossRef] [PubMed]
  22. Hui, E.; Cheung, J.; Zhu, J.; Su, X.; Taylor, M.J.; Wallweber, H.A.; Sasmal, D.K.; Huang, J.; Kim, J.M.; Mellman, I.; et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 2017, 355, 1428–1433. [Google Scholar] [CrossRef] [PubMed]
  23. Smith-Garvin, J.E.; Koretzky, G.A.; Jordan, M.S. T Cell Activation. Annu. Rev. Immunol. 2009, 27, 591–619. [Google Scholar] [CrossRef] [PubMed]
  24. Herbst, R.S.; Baas, P.; Kim, D.-W.; Felip, E.; Perez-Gracia, J.L.; Han, J.-Y.; Molina, J.; Kim, J.-H.; Arvis, C.D.; Ahn, M.-J.; et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): A randomised controlled trial. Lancet 2016, 387, 1540–1550. [Google Scholar] [CrossRef]
  25. Burtness, B.; Rischin, D.; Greil, R.; Soulières, D.; Tahara, M.; de Castro, G., Jr.; Psyrri, A.; Brana, I.; Basté, N.; Neupane, P.; et al. Pembrolizumab Alone or with Chemotherapy for Recurrent/Metastatic Head and Neck Squamous Cell Carcinoma in KEYNOTE-048: Subgroup Analysis by Programmed Death Ligand-1 Combined Positive Score. J. Clin. Oncol. 2022. [Google Scholar] [CrossRef]
  26. Atkins, M.B.; Jegede, O.A.; Haas, N.B.; McDermott, D.F.; Bilen, M.A.; Stein, M.; Sosman, J.A.; Alter, R.; Plimack, E.R.; Ornstein, M.; et al. Phase II Study of Nivolumab and Salvage Nivolumab/Ipilimumab in Treatment-Naive Patients with Advanced Clear Cell Renal Cell Carcinoma (HCRN GU16-260-Cohort A). J. Clin. Oncol. 2022, 40, 288. [Google Scholar] [CrossRef]
  27. Davis, A.A.; Patel, V.G. The role of PD-L1 expression as a predictive biomarker: An analysis of all US Food and Drug Administration (FDA) approvals of immune checkpoint inhibitors. J. Immunother. Cancer 2019, 7, 278. [Google Scholar] [CrossRef]
  28. Binnewies, M.; Roberts, E.W.; Kersten, K.; Chan, V.; Fearon, D.F.; Merad, M.; Coussens, L.M.; Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Hedrick, C.C.; et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat. Med. 2018, 24, 541–550. [Google Scholar] [CrossRef]
  29. McGranahan, N.; Swanton, C. Biological and Therapeutic Impact of Intratumor Heterogeneity in Cancer Evolution. Cancer Cell 2015, 27, 15–26. [Google Scholar] [CrossRef] [Green Version]
  30. Dagogo-Jack, I.; Shaw, A.T. Tumour heterogeneity and resistance to cancer therapies. Nat. Rev. Clin. Oncol. 2018, 15, 81–94. [Google Scholar] [CrossRef]
  31. Riaz, N.; Havel, J.; Makarov, V.; Desrichard, A.; Urba, W.J.; Sims, J.S.; Hodi, F.S.; Martín-Algarra, S.; Mandal, R.; Sharfman, W.H.; et al. Tumor and Microenvironment Evolution during Immunotherapy with Nivolumab. Cell 2017, 171, 934–949.e16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Girolami, I.; Pantanowitz, L.; Barberis, M.; Paolino, G.; Brunelli, M.; Vigliar, E.; Munari, E.; Satturwar, S.; Troncone, G.; Eccher, A. Challenges facing pathologists evaluating PD-L1 in head & neck squamous cell carcinoma. J. Oral Pathol. Med. 2021, 50, 864–873. [Google Scholar] [CrossRef] [PubMed]
  33. Yu, H.; Boyle, T.A.; Zhou, C.; Rimm, D.L.; Hirsch, F.R. PD-L1 Expression in Lung Cancer. J. Thorac. Oncol. 2016, 11, 964–975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Emancipator, K.; Huang, L.; Aurora-Garg, D.; Bal, T.; Cohen, E.E.W.; Harrington, K.; Soulières, D.; Le Tourneau, C.; Licitra, L.; Burtness, B.; et al. Comparing programmed death ligand 1 scores for predicting pembrolizumab efficacy in head and neck cancer. Mod. Pathol. 2021, 34, 532–541. [Google Scholar] [CrossRef]
  35. Russell-Goldman, E.; Kravets, S.; Dahlberg, S.; Sholl, L.M.; Vivero, M. Cytologic-histologic correlation of programmed death-ligand 1 immunohistochemistry in lung carcinomas. Cancer Cytopathol. 2018, 126, 253–263. [Google Scholar] [CrossRef] [Green Version]
  36. Satturwar, S.; Girolami, I.; Munari, E.; Ciompi, F.; Eccher, A.; Pantanowitz, L. Program death ligand-1 immunocytochemistry in lung cancer cytological samples: A systematic review. Diagn. Cytopathol. 2022, 50, 313–323. [Google Scholar] [CrossRef]
  37. Berry, S.; Giraldo, N.A.; Green, B.F.; Cottrell, T.R.; Stein, J.E.; Engle, E.L.; Xu, H.; Ogurtsova, A.; Roberts, C.; Wang, D.; et al. Analysis of multispectral imaging with the AstroPath platform informs efficacy of PD-1 blockade. Science 2021, 372, eaba2609. [Google Scholar] [CrossRef]
  38. Heitzer, E.; Haque, I.S.; Roberts, C.E.S.; Speicher, M.R. Current and future perspectives of liquid biopsies in genomics-driven oncology. Nat. Rev. Genet. 2019, 20, 71–88. [Google Scholar] [CrossRef]
  39. Hiam-Galvez, K.J.; Allen, B.M.; Spitzer, M.H. Systemic immunity in cancer. Nat. Cancer 2021, 21, 345–359. [Google Scholar] [CrossRef]
  40. Lucibello, G.; Mograbi, B.; Milano, G.; Hofman, P.; Brest, P. PD-L1 regulation revisited: Impact on immunotherapeutic strategies. Trends Mol. Med. 2021, 27, 868–881. [Google Scholar] [CrossRef]
  41. Zerdes, I.; Matikas, A.; Bergh, J.; Rassidakis, G.Z.; Foukakis, T. Genetic, transcriptional and post-translational regulation of the programmed death protein ligand 1 in cancer: Biology and clinical correlations. Oncogene 2018, 37, 4639–4661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Keir, M.E.; Butte, M.J.; Freeman, G.J.; Sharpe, A.H. PD-1 and Its Ligands in Tolerance and Immunity. Annu. Rev. Immunol. 2008, 26, 677–704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Hu, X.; Lin, Z.; Wang, Z.; Zhou, Q. Emerging role of PD-L1 modification in cancer immunotherapy. Am. J. Cancer Res. 2021, 11, 3832–3840. [Google Scholar] [PubMed]
  44. Mereiter, S.; Balmaña, M.; Campos, D.; Gomes, J.; Reis, C.A. Glycosylation in the era of cancer-targeted therapy: Where are we heading? Cancer Cell 2019, 36, 6–16. [Google Scholar] [CrossRef] [PubMed]
  45. Li, C.-W.; Lim, S.-O.; Xia, W.; Lee, H.-H.; Chan, L.-C.; Kuo, C.-W.; Khoo, K.-H.; Chang, S.-S.; Cha, J.-H.; Kim, T.; et al. Glycosylation and stabilization of programmed death ligand-1 suppresses T-cell activity. Nat. Commun. 2016, 7, 12632. [Google Scholar] [CrossRef] [Green Version]
  46. Gou, Q.; Dong, C.; Xu, H.; Khan, B.; Jin, J.; Liu, Q.; Shi, J.; Hou, Y. PD-L1 degradation pathway and immunotherapy for cancer. Cell Death Dis. 2020, 11, 955. [Google Scholar] [CrossRef]
  47. Cha, J.-H.; Chan, L.-C.; Li, C.-W.; Hsu, J.L.; Hung, M.-C. Mechanisms Controlling PD-L1 Expression in Cancer. Mol. Cell 2019, 76, 359–370. [Google Scholar] [CrossRef]
  48. Centanni, M.; Moes, D.J.A.R.; Trocóniz, I.F.; Ciccolini, J.; van Hasselt, J.G.C. Clinical Pharmacokinetics and Pharmacodynamics of Immune Checkpoint Inhibitors. Clin. Pharmacokinet. 2019, 58, 835–857. [Google Scholar] [CrossRef] [Green Version]
  49. Sun, C.; Mezzadra, R.; Schumacher, T.N. Regulation and Function of the PD-L1 Checkpoint. Immunity 2018, 48, 434–452. [Google Scholar] [CrossRef] [Green Version]
  50. Huang, R.S.P.; Haberberger, J.; Severson, E.; Duncan, D.L.; Hemmerich, A.; Edgerly, C.; Ferguson, N.L.; Williams, E.; Elvin, J.; Vergilio, J.-A.; et al. A pan-cancer analysis of PD-L1 immunohistochemistry and gene amplification, tumor mutation burden and microsatellite instability in 48,782 cases. Mod. Pathol. 2021, 34, 252–263. [Google Scholar] [CrossRef]
  51. Goodman, A.; Patel, S.P.; Kurzrock, R. PD-1-PD-L1 immune-checkpoint blockade in B-cell lymphomas. Nat. Rev. Clin. Oncol. 2017, 14, 203–220. [Google Scholar] [CrossRef] [PubMed]
  52. Roemer, M.G.M.; Advani, R.H.; Ligon, A.H.; Natkunam, Y.; Redd, R.A.; Homer, H.; Connelly, C.F.; Sun, H.H.; Daadi, S.E.; Freeman, G.J.; et al. PD-L1 and PD-L2 Genetic Alterations Define Classical Hodgkin Lymphoma and Predict Outcome. J. Clin. Oncol. 2016, 34, 2690–2697. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Goodman, A.M.; Piccioni, D.; Kato, S.; Boichard, A.; Wang, H.-Y.; Frampton, G.; Lippman, S.M.; Connelly, C.; Fabrizio, D.; Miller, V.; et al. Prevalence of PDL1 Amplification and Preliminary Response to Immune Checkpoint Blockade in Solid Tumors. JAMA Oncol. 2018, 4, 1237–1244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Kornepati, A.V.R.; Vadlamudi, R.K.; Curiel, T.J. Programmed death ligand 1 signals in cancer cells. Nat. Cancer 2022, 22, 174–189. [Google Scholar] [CrossRef] [PubMed]
  55. Dong, P.; Xiong, Y.; Yue, J.; Hanley, S.J.B.; Watari, H. Tumor-Intrinsic PD-L1 Signaling in Cancer Initiation, Development and Treatment: Beyond Immune Evasion. Front. Oncol. 2018, 8, 386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Sugiura, D.; Maruhashi, T.; Okazaki, I.-M.; Shimizu, K.; Maeda, T.K.; Takemoto, T.; Okazaki, T. Restriction of PD-1 function by cis -PD-L1/CD80 interactions is required for optimal T cell responses. Science 2019, 364, 558–566. [Google Scholar] [CrossRef]
  57. Zhao, Y.; Harrison, D.L.; Song, Y.; Ji, J.; Huang, J.; Hui, E. Antigen-Presenting Cell-Intrinsic PD-1 Neutralizes PD-L1 in cis to Attenuate PD-1 Signaling in T Cells. Cell Rep. 2018, 24, 379–390.e6. [Google Scholar] [CrossRef] [Green Version]
  58. Orme, J.J.; Jazieh, K.A.; Xie, T.; Harrington, S.; Liu, X.; Ball, M.; Madden, B.; Charlesworth, M.C.; Azam, T.U.; Lucien, F.; et al. ADAM10 and ADAM17 cleave PD-L1 to mediate PD-(L)1 inhibitor resistance. OncoImmunology 2020, 9, 1744980. [Google Scholar] [CrossRef] [Green Version]
  59. Romero, Y.; Wise, R.; Zolkiewska, A. Proteolytic processing of PD-L1 by ADAM proteases in breast cancer cells. Cancer Immunol. Immunother. 2020, 69, 43–55. [Google Scholar] [CrossRef]
  60. Xiong, W.; Gao, Y.; Wei, W.; Zhang, J. Extracellular and nuclear PD-L1 in modulating cancer immunotherapy. Trends Cancer 2021, 7, 837–846. [Google Scholar] [CrossRef]
  61. Zhou, J.; Mahoney, K.M.; Giobbie-Hurder, A.; Zhao, F.; Lee, S.; Liao, X.; Rodig, S.; Li, J.; Wu, X.; Butterfield, L.H.; et al. Soluble PD-L1 as a Biomarker in Malignant Melanoma Treated with Checkpoint Blockade. Cancer Immunol. Res. 2017, 5, 480–492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Sagawa, R.; Sakata, S.; Gong, B.; Seto, Y.; Takemoto, A.; Takagi, S.; Ninomiya, H.; Yanagitani, N.; Nakao, M.; Mun, M.; et al. Soluble PD-L1 works as a decoy in lung cancer immunotherapy via alternative polyadenylation. JCI Insight 2022, 7, e153323. [Google Scholar] [CrossRef] [PubMed]
  63. Mildner, F.; Sopper, S.; Amann, A.; Pircher, A.; Pall, G.; Köck, S.; Naismith, E.; Wolf, D.; Gamerith, G. Systematic review: Soluble immunological biomarkers in advanced non-small-cell lung cancer (NSCLC). Crit. Rev. Oncol. 2020, 153, 102948. [Google Scholar] [CrossRef] [PubMed]
  64. Bailly, C.; Thuru, X.; Quesnel, B. Soluble Programmed Death Ligand-1 (sPD-L1): A Pool of Circulating Proteins Implicated in Health and Diseases. Cancers 2021, 13, 3034. [Google Scholar] [CrossRef] [PubMed]
  65. Ugurel, S.; Schadendorf, D.; Horny, K.; Sucker, A.; Schramm, S.; Utikal, J.; Pföhler, C.; Herbst, R.; Schilling, B.; Blank, C.; et al. Elevated baseline serum PD-1 or PD-L1 predicts poor outcome of PD-1 inhibition therapy in metastatic melanoma. Ann. Oncol. 2020, 31, 144–152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Del Re, M.; Marconcini, R.; Pasquini, G.; Rofi, E.; Vivaldi, C.; Bloise, F.; Restante, G.; Arrigoni, E.; Caparello, C.; Bianco, M.G.; et al. PD-L1 mRNA expression in plasma-derived exosomes is associated with response to anti-PD-1 antibodies in melanoma and NSCLC. Br. J. Cancer 2018, 118, 820–824. [Google Scholar] [CrossRef] [Green Version]
  67. Grossman, J.E.; Vasudevan, D.; Joyce, C.E.; Hildago, M. Is PD-L1 a consistent biomarker for anti-PD-1 therapy? The model of balstilimab in a virally-driven tumor. Oncogene 2021, 40, 1393–1395. [Google Scholar] [CrossRef]
  68. Nicolazzo, C.; Raimondi, C.; Mancini, M.; Caponnetto, S.; Gradilone, A.; Gandini, O.; Mastromartino, M.; Del Bene, G.; Prete, A.A.; Longo, F.; et al. Monitoring PD-L1 positive circulating tumor cells in non-small cell lung cancer patients treated with the PD-1 inhibitor Nivolumab. Sci. Rep. 2016, 6, 31726. [Google Scholar] [CrossRef]
  69. Shimada, Y.; Matsubayashi, J.; Kudo, Y.; Maehara, S.; Takeuchi, S.; Hagiwara, M.; Kakihana, M.; Ohira, T.; Nagao, T.; Ikeda, N. Serum-derived exosomal PD-L1 expression to predict anti-PD-1 response and in patients with non-small cell lung cancer. Sci. Rep. 2021, 11, 7830. [Google Scholar] [CrossRef]
  70. Strati, A.; Koutsodontis, G.; Papaxoinis, G.; Angelidis, I.; Zavridou, M.; Economopoulou, P.; Kotsantis, I.; Avgeris, M.; Mazel, M.; Perisanidis, C.; et al. Prognostic significance of PD-L1 expression on circulating tumor cells in patients with head and neck squamous cell carcinoma. Ann. Oncol. 2017, 28, 1923–1933. [Google Scholar] [CrossRef]
  71. Hamilton, G.; Moser, D.; Hochmair, M. Metastasis: Circulating Tumor Cells in Small Cell Lung Cancer. Trends Cancer 2016, 2, 159–160. [Google Scholar] [CrossRef] [PubMed]
  72. Raimondi, C.; Carpino, G.; Nicolazzo, C.; Gradilone, A.; Gianni, W.; Gelibter, A.; Gaudio, E.; Cortesi, E.; Gazzaniga, P. PD-L1 and epithelial-mesenchymal transition in circulating tumor cells from non-small cell lung cancer patients: A molecular shield to evade immune system? Oncoimmunology 2017, 6, e1315488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Mansfield, A.S.; Aubry, M.C.; Moser, J.C.; Harrington, S.M.; Dronca, R.S.; Park, S.S.; Dong, H. Temporal and spatial discordance of programmed cell death-ligand 1 expression and lymphocyte tumor infiltration between paired primary lesions and brain metastases in lung cancer. Ann. Oncol. 2016, 27, 1953–1958. [Google Scholar] [CrossRef] [PubMed]
  74. Madore, J.; Strbenac, D.; Vilain, R.; Menzies, A.M.; Yang, J.Y.H.; Thompson, J.F.; Long, G.V.; Mann, G.J.; Scolyer, R.A.; Wilmott, J.S. PD-L1 Negative Status is Associated with Lower Mutation Burden, Differential Expression of Immune-Related Genes, and Worse Survival in Stage III Melanoma. Clin. Cancer Res. 2016, 22, 3915–3923. [Google Scholar] [CrossRef] [Green Version]
  75. Ahn, J.C.; Teng, P.C.; Chen, P.J.; Posadas, E.; Tseng, H.R.; Lu, S.C.; Yang, J.D. Detection of circulating tumor cells and their implications as a novel biomarker for diagnosis, prognostication, and therapeutic monitoring in hepatocellular carcinoma. Hepatology 2021, 73, 422–436. [Google Scholar] [CrossRef]
  76. Tamminga, M.; de Wit, S.; Hiltermann, T.J.N.; Timens, W.; Schuuring, E.; Terstappen, L.; Groen, H.J. Circulating tumor cells in advanced non-small cell lung cancer patients are associated with worse tumor response to checkpoint inhibitors. J. Immunother. Cancer 2019, 7, 173. [Google Scholar] [CrossRef]
  77. Ilié, M.; Szafer-Glusman, E.; Hofman, V.; Chamorey, E.; Lalvée, S.; Selva, E.; Leroy, S.; Marquette, C.-H.; Kowanetz, M.; Hedge, P.; et al. Detection of PD-L1 in circulating tumor cells and white blood cells from patients with advanced non-small-cell lung cancer. Ann. Oncol. 2018, 29, 193–199. [Google Scholar] [CrossRef]
  78. Khattak, M.A.; Reid, A.; Freeman, J.; Pereira, M.; McEvoy, A.; Lo, J.; Frank, M.H.; Meniawy, T.; Didan, A.; Spencer, I.; et al. PD-L1 Expression on Circulating Tumor Cells May Be Predictive of Response to Pembrolizumab in Advanced Melanoma: Results from a Pilot Study. Oncol. 2019, 25, e520–e527. [Google Scholar] [CrossRef] [Green Version]
  79. Kloten, V.; Lampignano, R.; Krahn, T.; Schlange, T. Circulating Tumor Cell PD-L1 Expression as Biomarker for Therapeutic Efficacy of Immune Checkpoint Inhibition in NSCLC. Cells 2019, 8, 809. [Google Scholar] [CrossRef] [Green Version]
  80. Boffa, D.J.; Graf, R.P.; Salazar, M.C.; Hoag, J.; Lu, D.; Krupa, R.; Louw, J.; Dugan, L.; Wang, Y.; Landers, M.; et al. Cellular Expression of PD-L1 in the Peripheral Blood of Lung Cancer Patients is Associated with Worse Survival. Cancer Epidemiol. Biomark. Prev. 2017, 26, 1139–1145. [Google Scholar] [CrossRef] [Green Version]
  81. Guibert, N.; Delaunay, M.; Lusque, A.; Boubekeur, N.; Rouquette, I.; Clermont, E.; Mourlanette, J.; Gouin, S.; Dormoy, I.; Favre, G.; et al. PD-L1 expression in circulating tumor cells of advanced non-small cell lung cancer patients treated with nivolumab. Lung Cancer 2018, 120, 108–112. [Google Scholar] [CrossRef] [PubMed]
  82. Sinoquet, L.; Jacot, W.; Gauthier, L.; Pouderoux, S.; Viala, M.; Cayrefourcq, L.; Quantin, X.; Alix-Panabières, C. Programmed Cell Death Ligand 1-Expressing Circulating Tumor Cells: A New Prognostic Biomarker in Non-Small Cell Lung Cancer. Clin. Chem. 2021, 67, 1503–1512. [Google Scholar] [CrossRef] [PubMed]
  83. Brozos-Vázquez, E.M.; Díaz-Peña, R.; García-González, J.; León-Mateos, L.; Mondelo-Macía, P.; Peña-Chilet, M.; López-López, R. Immunotherapy in nonsmall-cell lung cancer: Current status and future prospects for liquid biopsy. Cancer Immunol. Immunother. 2021, 70, 1177–1188. [Google Scholar] [CrossRef] [PubMed]
  84. Palicelli, A.; Bonacini, M.; Croci, S.; Magi-Galluzzi, C.; Cañete-Portillo, S.; Chaux, A.; Bisagni, A.; Zanetti, E.; De Biase, D.; Melli, B. What Do We Have to Know about PD-L1 Expression in Prostate Cancer? A Systematic Literature Review. Part 2: Clinic–Pathologic Correlations. Cells 2021, 10, 3165. [Google Scholar] [CrossRef]
  85. Koh, Y.; Yagi, S.; Akamatsu, H.; Kanai, K.; Hayata, A.; Tokudome, N.; Akamatsu, K.; Higuchi, M.; Kanbara, H.; Nakanishi, M.; et al. Heterogeneous Expression of Programmed Death Receptor-ligand 1 on Circulating Tumor Cells in Patients with Lung Cancer. Clin. Lung Cancer 2019, 20, 270–277.e1. [Google Scholar] [CrossRef] [Green Version]
  86. Tanaka, F.; Yoneda, K.; Kondo, N.; Hashimoto, M.; Takuwa, T.; Matsumoto, S.; Okumura, Y.; Rahman, S.; Tsubota, N.; Tsujimura, T.; et al. Circulating Tumor Cell as a Diagnostic Marker in Primary Lung Cancer. Clin. Cancer Res. 2009, 15, 6980–6986. [Google Scholar] [CrossRef] [Green Version]
  87. Chen, Y.-L.; Huang, W.-C.; Lin, F.-M.; Hsieh, H.B.; Hsieh, C.-H.; Hsieh, R.K.; Chen, K.-W.; Yen, M.-H.; Lee, J.; Su, S.; et al. Novel circulating tumor cell-based blood test for the assessment of PD-L1 protein expression in treatment-naïve, newly diagnosed patients with non-small cell lung cancer. Cancer Immunol. Immunother. 2019, 68, 1087–1094. [Google Scholar] [CrossRef] [Green Version]
  88. Satelli, A.; Batth, I.S.; Brownlee, Z.; Rojas, C.; Meng, Q.H.; Kopetz, S.; Li, S. Potential role of nuclear PD-L1 expression in cell-surface vimentin positive circulating tumor cells as a prognostic marker in cancer patients. Sci. Rep. 2016, 6, 28910. [Google Scholar] [CrossRef]
  89. Schott, D.S.; Pizon, M.; Pachmann, U.; Pachmann, K. Sensitive detection of PD-L1 expression on circulating epithelial tumor cells (CETCs) could be a potential biomarker to select patients for treatment with PD-1/PD-L1 inhibitors in early and metastatic solid tumors. Oncotarget 2017, 8, 72755. [Google Scholar] [CrossRef] [Green Version]
  90. Bankó, P.; Lee, S.Y.; Nagygyörgy, V.; Zrínyi, M.; Chae, C.H.; Cho, D.H.; Telekes, A. Technologies for circulating tumor cell separation from whole blood. J. Hematol. Oncol. 2019, 12, 48. [Google Scholar] [CrossRef] [Green Version]
  91. Liu, B.; Dong, K.; Dong, R. Techniques and Thresholds for Quantifying Circulating Tumor Cells in Breast Cancer. JAMA Oncol. 2019, 5, 573. [Google Scholar] [CrossRef] [PubMed]
  92. Allard, W.J.; Matera, J.; Miller, M.C.; Repollet, M.; Connelly, M.C.; Rao, C.; Tibbe, A.G.J.; Uhr, J.W.; Terstappen, L.W.M.M. Tumor cells circulate in the peripheral blood of all major carcinomas but not in healthy subjects or patients with nonmalignant diseases. Clin. Cancer Res. 2004, 10, 6897–6904. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Kilgour, E.; Rothwell, D.G.; Brady, G.; Dive, C. Liquid Biopsy-Based Biomarkers of Treatment Response and Resistance. Cancer Cell 2020, 37, 485–495. [Google Scholar] [CrossRef] [PubMed]
  94. Andree, K.C.; Van Dalum, G.; Terstappen, L.W.M.M. Challenges in circulating tumor cell detection by the CellSearch system. Mol. Oncol. 2016, 10, 395–407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Ferreira, M.M.; Ramani, V.C.; Jeffrey, S.S. Circulating tumor cell technologies. Mol. Oncol. 2016, 10, 374–394. [Google Scholar] [CrossRef] [Green Version]
  96. Gorges, T.M.; Tinhofer, I.; Drosch, M.; Röse, L.; Zollner, T.M.; Krahn, T.; Von Ahsen, O. Circulating tumour cells escape from EpCAM-based detection due to epithelial-to-mesenchymal transition. BMC Cancer 2012, 12, 178. [Google Scholar] [CrossRef] [Green Version]
  97. Alix-Panabières, C.; Pantel, K. Challenges in circulating tumour cell research. Nat. Cancer 2014, 14, 623–631. [Google Scholar] [CrossRef]
  98. Alix-Panabières, C.; Pantel, K. Liquid Biopsy: From Discovery to Clinical Application. Cancer Discov. 2021, 11, 858–873. [Google Scholar] [CrossRef]
  99. 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]
  100. Mazieres, J.; Rittmeyer, A.; Gadgeel, S.; Hida, T.; Gandara, D.R.; Cortinovis, D.L.; Barlesi, F.; Yu, W.; Matheny, C.; Ballinger, M.; et al. Atezolizumab Versus Docetaxel in Pretreated Patients With NSCLC: Final Results from the Randomized Phase 2 POPLAR and Phase 3 OAK Clinical Trials. J. Thorac. Oncol. 2021, 16, 140–150. [Google Scholar] [CrossRef]
  101. Boman, C.; Zerdes, I.; Mårtensson, K.; Bergh, J.; Foukakis, T.; Valachis, A.; Matikas, A. Discordance of PD-L1 status between primary and metastatic breast cancer: A systematic review and meta-analysis. Cancer Treat. Rev. 2021, 99, 102257. [Google Scholar] [CrossRef] [PubMed]
  102. Rozenblit, M.; Huang, R.; Danziger, N.; Hegde, P.; Alexander, B.; Ramkissoon, S.; Blenman, K.; Ross, J.S.; Rimm, D.L.; Pusztai, L. Comparison of PD-L1 protein expression between primary tumors and metastatic lesions in triple negative breast cancers. J. Immunother. Cancer 2020, 8, e001558. [Google Scholar] [CrossRef] [PubMed]
  103. Aceto, N.; Bardia, A.; Miyamoto, D.T.; Donaldson, M.C.; Wittner, B.S.; Spencer, J.A.; Yu, M.; Pely, A.; Engstrom, A.; Zhu, H.; et al. Circulating Tumor Cell Clusters Are Oligoclonal Precursors of Breast Cancer Metastasis. Cell 2014, 158, 1110–1122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Lee, H.-H.; Wang, Y.-N.; Xia, W.; Chen, C.-H.; Rau, K.-M.; Ye, L.; Wei, Y.; Chou, C.-K.; Wang, S.-C.; Yan, M.; et al. Removal of N-Linked Glycosylation Enhances PD-L1 Detection and Predicts Anti-PD-1/PD-L1 Therapeutic Efficacy. Cancer Cell 2019, 36, 168–178.e4. [Google Scholar] [CrossRef] [PubMed]
  105. Sidaway, P. Deglycosylated PD-L1 might be a better biomarker. Nat. Rev. Clin. Oncol. 2019, 16, 592. [Google Scholar] [CrossRef]
  106. Mei, J.; Xu, J.; Yang, X.; Gu, D.; Zhou, W.; Wang, H.; Liu, C. A comparability study of natural and deglycosylated PD-L1 levels in lung cancer: Evidence from immunohistochemical analysis. Mol. Cancer 2021, 20, 11. [Google Scholar] [CrossRef]
  107. Liu, X.; Taftaf, R.; Kawaguchi, M.; Chang, Y.-F.; Chen, W.; Entenberg, D.; Zhang, Y.; Gerratana, L.; Huang, S.; Patel, D.B.; et al. Homophilic CD44 Interactions Mediate Tumor Cell Aggregation and Polyclonal Metastasis in Patient-Derived Breast Cancer Models. Cancer Discov. 2019, 9, 96–113. [Google Scholar] [CrossRef] [Green Version]
  108. Schuster, E.; Taftaf, R.; Reduzzi, C.; Albert, M.K.; Romero-Calvo, I.; Liu, H. Better together: Circulating tumor cell clustering in metastatic cancer. Trends Cancer 2021, 7, 1020–1032. [Google Scholar] [CrossRef]
  109. Jansson, S.; Bendahl, P.-O.; Larsson, A.-M.; Aaltonen, K.E.; Rydén, L. Prognostic impact of circulating tumor cell apoptosis and clusters in serial blood samples from patients with metastatic breast cancer in a prospective observational cohort. BMC Cancer 2016, 16, 433. [Google Scholar] [CrossRef] [Green Version]
  110. Szczerba, B.M.; Castro-Giner, F.; Vetter, M.; Krol, I.; Gkountela, S.; Landin, J.; Scheidmann, M.C.; Donato, C.; Scherrer, R.; Singer, J.; et al. Neutrophils escort circulating tumour cells to enable cell cycle progression. Nature 2019, 566, 553–557. [Google Scholar] [CrossRef]
  111. Manjunath, Y.; Upparahalli, S.V.; Suvilesh, K.N.; Avella, D.M.; Kimchi, E.T.; Staveley-O’Carroll, K.F.; Li, G.; Kaifi, J.T. Circulating tumor cell clusters are a potential biomarker for detection of non-small cell lung cancer. Lung Cancer 2019, 134, 147–150. [Google Scholar] [CrossRef] [PubMed]
  112. Saini, M.; Szczerba, B.M.; Aceto, N. Circulating Tumor Cell-Neutrophil Tango along the Metastatic Process. Cancer Res. 2019, 79, 6067–6073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Nishimura, C.D.; Pulanco, M.C.; Cui, W.; Lu, L.; Zang, X. PD-L1 and B7-1 Cis-Interaction: New Mechanisms in Immune Checkpoints and Immunotherapies. Trends Mol. Med. 2021, 27, 207–219. [Google Scholar] [CrossRef] [PubMed]
  114. VanderWalde, A.; Spetzler, D.; Xiao, N.; Gatalica, Z.; Marshall, J. Microsatellite instability status determined by next-generation sequencing and compared with PD-L1 and tumor mutational burden in 11,348 patients. Cancer Med. 2018, 7, 746–756. [Google Scholar] [CrossRef] [Green Version]
  115. Yarchoan, M.; Albacker, L.A.; Hopkins, A.C.; Montesion, M.; Murugesan, K.; Vithayathil, T.T.; Zaidi, N.; Azad, N.S.; Laheru, D.A.; Frampton, G.M.; et al. PD-L1 expression and tumor mutational burden are independent biomarkers in most cancers. JCI Insight 2019, 4, e126908. [Google Scholar] [CrossRef] [Green Version]
  116. Goodman, A.M.; Kato, S.; Bazhenova, L.; Patel, S.P.; Frampton, G.M.; Miller, V.; Stephens, P.J.; Daniels, G.A.; Kurzrock, R. Tumor Mutational Burden as an Independent Predictor of Response to Immunotherapy in Diverse Cancers. Mol. Cancer Ther. 2017, 16, 2598–2608. [Google Scholar] [CrossRef] [Green Version]
  117. Marabelle, A.; Fakih, M.; Lopez, J.; Shah, M.; Shapira-Frommer, R.; Nakagawa, K.; Chung, H.C.; Kindler, H.L.; Lopez-Martin, J.A.; Miller, W.H., Jr.; et al. Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: Prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol. 2020, 21, 1353–1365. [Google Scholar] [CrossRef]
  118. Inoue, Y.; Yoshimura, K.; Nishimoto, K.; Inui, N.; Karayama, M.; Yasui, H.; Hozumi, H.; Suzuki, Y.; Furuhashi, K.; Fujisawa, T.; et al. Evaluation of Programmed Death Ligand 1 (PD-L1) Gene Amplification and Response to Nivolumab Monotherapy in Non–small Cell Lung Cancer. JAMA Netw. Open 2020, 3, e2011818. [Google Scholar] [CrossRef]
  119. Lamberti, G.; Spurr, L.F.; Li, Y.; Ricciuti, B.; Recondo, G.; Umeton, R.; Nishino, M.; Sholl, L.M.; Meyerson, M.L.; Cherniack, A.D.; et al. Clinicopathological and genomic correlates of programmed cell death ligand 1 (PD-L1) expression in nonsquamous non-small-cell lung cancer. Ann. Oncol. 2020, 31, 807–814. [Google Scholar] [CrossRef]
  120. Poudineh, M.; Sargent, E.H.; Pantel, K.; Kelley, S.O. Profiling circulating tumour cells and other biomarkers of invasive cancers. Nat. Biomed. Eng. 2018, 2, 72–84. [Google Scholar] [CrossRef]
  121. Nagrath, S.; Sequist, L.V.; Maheswaran, S.; Bell, D.W.; Irimia, D.; Ulkus, L.; Smith, M.R.; Kwak, E.L.; Digumarthy, S.; Muzikansky, A.; et al. Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature 2007, 450, 1235–1239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Gandara, D.R.; Paul, S.M.; Kowanetz, M.; Schleifman, E.; Zou, W.; Li, Y.; Rittmeyer, A.; Fehrenbacher, L.; Otto, G.; Malboeuf, C.; et al. Blood-based tumor mutational burden as a predictor of clinical benefit in non-small-cell lung cancer patients treated with atezolizumab. Nat. Med. 2018, 24, 1441–1448. [Google Scholar] [CrossRef] [PubMed]
  123. Keller, L.; Pantel, K. Unravelling tumour heterogeneity by single-cell profiling of circulating tumour cells. Nat. Cancer 2019, 19, 553–567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Labib, M.; Kelley, S.O. Single-cell analysis targeting the proteome. Nat. Rev. Chem. 2020, 4, 143–158. [Google Scholar] [CrossRef]
  125. Krebs, M.G.; Metcalf, R.L.; Carter, L.; Brady, G.; Blackhall, F.H.; Dive, C. Molecular analysis of circulating tumour cells—Biology and biomarkers. Nat. Rev. Clin. Oncol. 2014, 11, 129–144. [Google Scholar] [CrossRef]
  126. Pal, A.; Shinde, R.; Miralles, M.S.; Workman, P.; de Bono, J. Applications of liquid biopsy in the Pharmacological Audit Trail for anticancer drug development. Nat. Rev. Clin. Oncol. 2021, 18, 454–467. [Google Scholar] [CrossRef]
  127. Salmon, H.; Remark, R.; Gnjatic, S.; Merad, M. Host tissue determinants of tumour immunity. Nat. Cancer 2019, 19, 215–227. [Google Scholar] [CrossRef]
  128. Gonzalez, H.; Hagerling, C.; Werb, Z. Roles of the immune system in cancer: From tumor initiation to metastatic progression. Genes Dev. 2018, 32, 1267–1284. [Google Scholar] [CrossRef] [Green Version]
  129. Guo, X.; Zhang, Y.; Zheng, L.; Zheng, C.; Song, J.; Zhang, Q.; Kang, B.; Liu, Z.; Jin, L.; Xing, R.; et al. Global characterization of T cells in non-small-cell lung cancer by single-cell sequencing. Nat. Med. 2018, 24, 978–985. [Google Scholar] [CrossRef]
  130. Lavin, Y.; Kobayashi, S.; Leader, A.; Amir, E.D.; Elefant, N.; Bigenwald, C.; Remark, R.; Sweeney, R.; Becker, C.D.; Levine, J.H.; et al. Innate Immune Landscape in Early Lung Adenocarcinoma by Paired Single-Cell Analyses. Cell 2017, 169, 750–765.e17. [Google Scholar] [CrossRef] [Green Version]
  131. Gide, T.N.; Quek, C.; Menzies, A.M.; Tasker, A.T.; Shang, P.; Holst, J.; Madore, J.; Lim, S.Y.; Velickovic, R.; Wongchenko, M.; et al. Distinct Immune Cell Populations Define Response to Anti-PD-1 Monotherapy and Anti-PD-1/Anti-CTLA-4 Combined Therapy. Cancer Cell 2019, 35, 238–255.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Shaked, Y. The pro-tumorigenic host response to cancer therapies. Nat. Cancer 2019, 19, 667–685. [Google Scholar] [CrossRef] [PubMed]
  133. Bruni, D.; Angell, H.K.; Galon, J. The immune contexture and Immunoscore in cancer prognosis and therapeutic efficacy. Nat. Cancer 2020, 20, 662–680. [Google Scholar] [CrossRef] [PubMed]
  134. Johnson, R.M.G.; Dong, H. Functional Expression of Programmed Death-Ligand 1 (B7-H1) by Immune Cells and Tumor Cells. Front. Immunol. 2017, 8, 961. [Google Scholar] [CrossRef]
  135. Taube, J.M.; Klein, A.; Brahmer, J.R.; Xu, H.; Pan, X.; Kim, J.H.; Chen, L.; Pardoll, D.M.; Topalian, S.L.; Anders, R.A. Association of PD-1, PD-1 Ligands, and Other Features of the Tumor Immune Microenvironment with Response to Anti–PD-1 Therapy. Clin. Cancer Res. 2014, 20, 5064–5074. [Google Scholar] [CrossRef] [Green Version]
  136. Bergers, G.; Benjamin, L.E. Tumorigenesis and the angiogenic switch. Nat. Cancer 2003, 3, 401–410. [Google Scholar] [CrossRef]
  137. Farnsworth, R.H.; Lackmann, M.; Achen, M.G.; Stacker, S.A. Vascular remodeling in cancer. Oncogene 2013, 33, 3496–3505. [Google Scholar] [CrossRef] [Green Version]
  138. Padera, T.P.; Meijer, E.F.; Munn, L.L. The Lymphatic System in Disease Processes and Cancer Progression. Annu. Rev. Biomed. Eng. 2016, 18, 125–158. [Google Scholar] [CrossRef] [Green Version]
  139. Wu, T.D.; Madireddi, S.; de Almeida, P.E.; Banchereau, R.; Chen, Y.-J.J.; Chitre, A.S.; Chiang, E.Y.; Iftikhar, H.; O’Gorman, W.E.; Au-Yeung, A.; et al. Peripheral T cell expansion predicts tumour infiltration and clinical response. Nature 2020, 579, 274–278. [Google Scholar] [CrossRef]
  140. Spitzer, M.H.; Carmi, Y.; Reticker-Flynn, N.; Kwek, S.S.; Madhireddy, D.; Martins, M.M.; Gherardini, P.F.; Prestwood, T.R.; Chabon, J.; Bendall, S.C.; et al. Systemic Immunity Is Required for Effective Cancer Immunotherapy. Cell 2017, 168, 487–502.e15. [Google Scholar] [CrossRef] [Green Version]
  141. Lyon, A.R.; Yousaf, N.; Battisti, N.M.L.; Moslehi, J.; Larkin, J. Immune checkpoint inhibitors and cardiovascular toxicity. Lancet Oncol. 2018, 19, e447–e458. [Google Scholar] [CrossRef]
  142. Sullivan, R.J.; Weber, J.S. Immune-related toxicities of checkpoint inhibitors: Mechanisms and mitigation strategies. Nat. Rev. Drug Discov. 2021, 1–14. [Google Scholar] [CrossRef] [PubMed]
  143. Marin-Acevedo, J.A.; Chirila, R.M.; Dronca, R.S. Immune Checkpoint Inhibitor Toxicities. Mayo Clin. Proc. 2019, 94, 1321–1329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Valpione, S.; Galvani, E.; Tweedy, J.; Mundra, P.A.; Banyard, A.; Middlehurst, P.; Barry, J.; Mills, S.; Salih, Z.; Weightman, J.; et al. Immune awakening revealed by peripheral T cell dynamics after one cycle of immunotherapy. Nat. Cancer 2020, 1, 210–221. [Google Scholar] [CrossRef]
  145. Hernandez, C.; Arasanz, H.; Chocarro, L.; Bocanegra, A.; Zuazo, M.; Fernandez-Hinojal, G.; Blanco, E.; Vera, R.; Escors, D.; Kochan, G. Systemic Blood Immune Cell Populations as Biomarkers for the Outcome of Immune Checkpoint Inhibitor Therapies. Int. J. Mol. Sci. 2020, 21, 2411. [Google Scholar] [CrossRef] [Green Version]
  146. Bocanegra, A.; Fernandez-Hinojal, G.; Zuazo-Ibarra, M.; Arasanz, H.; Garcia-Granda, M.J.; Hernandez, C.; Ibañez, M.; Hernandez-Marin, B.; Martinez-Aguillo, M.; Lecumberri, M.J.; et al. PD-L1 Expression in Systemic Immune Cell Populations as a Potential Predictive Biomarker of Responses to PD-L1/PD-1 Blockade Therapy in Lung Cancer. Int. J. Mol. Sci. 2019, 20, 1631. [Google Scholar] [CrossRef] [Green Version]
  147. Engblom, C.; Pfirschke, C.; Pittet, M.J. The role of myeloid cells in cancer therapies. Nat. Rev. Cancer 2016, 16, 447–462. [Google Scholar] [CrossRef]
  148. Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Bronte, V. Coordinated regulation of myeloid cells by tumours. Nat. Rev. Immunol. 2012, 12, 253–268. [Google Scholar] [CrossRef] [Green Version]
  149. Peranzoni, E.; Ingangi, V.; Masetto, E.; Pinton, L.; Marigo, I. Myeloid Cells as Clinical Biomarkers for Immune Checkpoint Blockade. Front. Immunol. 2020, 11, 1590. [Google Scholar] [CrossRef]
  150. Moses, K.; Brandau, S. Human neutrophils: Their role in cancer and relation to myeloid-derived suppressor cells. Semin. Immunol. 2016, 28, 187–196. [Google Scholar] [CrossRef]
  151. Zhang, B.; Wang, Z.; Wu, L.; Zhang, M.; Li, W.; Ding, J.-H.; Zhu, J.; Wei, H.; Zhao, K. Circulating and Tumor-Infiltrating Myeloid-Derived Suppressor Cells in Patients with Colorectal Carcinoma. PLoS ONE 2013, 8, e57114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Elliott, L.A.; Doherty, G.; Sheahan, K.; Ryan, E.J. Human Tumor-Infiltrating Myeloid Cells: Phenotypic and Functional Diversity. Front. Immunol. 2017, 8, 86. [Google Scholar] [CrossRef] [PubMed]
  153. Siwicki, M.; Pittet, M.J. Versatile neutrophil functions in cancer. Semin Immunol. 2021, 101538. [Google Scholar]
  154. Shaul, M.E.; Fridlender, Z.G. Neutrophils as active regulators of the immune system in the tumor microenvironment. J. Leukoc. Biol. 2017, 102, 343–349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Kim, S.T.; Cristescu, R.; Bass, A.J.; Kim, K.-M.; Odegaard, J.I.; Kim, K.; Liu, X.Q.; Sher, X.; Jung, H.; Lee, M.; et al. Comprehensive molecular characterization of clinical responses to PD-1 inhibition in metastatic gastric cancer. Nat. Med. 2018, 24, 1449–1458. [Google Scholar] [CrossRef] [PubMed]
  156. Faget, J.; Peters, S.; Quantin, X.; Meylan, E.; Bonnefoy, N. Neutrophils in the era of immune checkpoint blockade. J. Immunother. Cancer 2021, 9, e002242. [Google Scholar] [CrossRef]
  157. Fukui, T.; Okuma, Y.; Nakahara, Y.; Otani, S.; Igawa, S.; Katagiri, M.; Mitsufuji, H.; Kubota, M.; Hiyoshi, Y.; Ishihara, M.; et al. Activity of Nivolumab and Utility of Neutrophil-to-Lymphocyte Ratio as a Predictive Biomarker for Advanced Non–Small-Cell Lung Cancer: A Prospective Observational Study. Clin. Lung Cancer 2018, 20, 208–214.e2. [Google Scholar] [CrossRef]
  158. Capone, M.; Giannarelli, D.; Mallardo, D.; Madonna, G.; Festino, L.; Grimaldi, A.M.; Vanella, V.; Simeone, E.; Paone, M.; Palmieri, G.; et al. Baseline neutrophil-to-lymphocyte ratio (NLR) and derived NLR could predict overall survival in patients with advanced melanoma treated with nivolumab. J. Immunother. Cancer 2018, 6, 74. [Google Scholar] [CrossRef] [Green Version]
  159. Valero, C.; Lee, M.; Hoen, D.; Weiss, K.; Kelly, D.W.; Adusumilli, P.S.; Paik, P.K.; Plitas, G.; Ladanyi, M.; Postow, M.A.; et al. Pretreatment neutrophil-to-lymphocyte ratio and mutational burden as biomarkers of tumor response to immune checkpoint inhibitors. Nat. Commun. 2021, 12, 729. [Google Scholar] [CrossRef]
  160. Sánchez-Gastaldo, A.; Muñoz-Fuentes, M.A.; Molina-Pinelo, S.; Alonso-García, M.; Boyero, L.; Bernabé-Caro, R. Correlation of peripheral blood biomarkers with clinical outcomes in NSCLC patients with high PD-L1 expression treated with pembrolizumab. Transl. Lung Cancer Res. 2021, 10, 2509–2522. [Google Scholar] [CrossRef]
  161. Hasegawa, T.; Yanagitani, N.; Utsumi, H.; Wakui, H.; Sakamoto, H.; Tozuka, T.; Yoshida, H.; Amino, Y.; Uematsu, S.; Yoshizawa, T.; et al. Association of High Neutrophil-to-Lymphocyte Ratio with Poor Outcomes of Pembrolizumab Therapy in High-PD-L1-expressing Non-small Cell Lung Cancer. Anticancer Res. 2019, 39, 6851–6857. [Google Scholar] [CrossRef]
  162. Park, W.; Kwon, D.; Saravia, D.; Desai, A.; Vargas, F.; El Dinali, M.; Warsch, J.; Elias, R.; Chae, Y.K.; Kim, D.W.; et al. Developing a Predictive Model for Clinical Outcomes of Advanced Non-Small Cell Lung Cancer Patients Treated with Nivolumab. Clin. Lung Cancer 2018, 19, 280–288.e4. [Google Scholar] [CrossRef] [PubMed]
  163. Ayers, K.L.; Ma, M.; Debussche, G.; Corrigan, D.; McCafferty, J.; Lee, K.; Newman, S.; Zhou, X.; Hirsch, F.R.; Mack, P.C.; et al. A composite biomarker of neutrophil-lymphocyte ratio and hemoglobin level correlates with clinical response to PD-1 and PD-L1 inhibitors in advanced non-small cell lung cancers. BMC Cancer 2021, 21, 441. [Google Scholar] [CrossRef] [PubMed]
  164. Naqash, A.R.; Stroud, C.R.G.; Butt, M.U.; Dy, G.K.; Hegde, A.; Muzaffar, M.; Yang, L.V.; Hafiz, M.; Cherry, C.R.; Walker, P.R. Co-relation of overall survival with peripheral blood-based inflammatory biomarkers in advanced stage non-small cell lung cancer treated with anti-programmed cell death-1 therapy: Results from a single institutional database. Acta Oncol. 2018, 57, 867–872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Soyano, A.E.; Dholaria, B.; Marin-Acevedo, J.A.; Diehl, N.; Hodge, D.; Luo, Y.; Manochakian, R.; Chumsri, S.; Adjei, A.; Knutson, K.L.; et al. Peripheral blood biomarkers correlate with outcomes in advanced non-small cell lung Cancer patients treated with anti-PD-1 antibodies. J. Immunother. Cancer 2018, 6, 129. [Google Scholar] [CrossRef] [PubMed]
  166. Ameratunga, M.; Chénard-Poirier, M.; Candilejo, I.M.; Pedregal, M.; Lui, A.; Dolling, D.; Aversa, C.; Garces, A.I.; Ang, J.E.; Banerji, U.; et al. Neutrophil-lymphocyte ratio kinetics in patients with advanced solid tumours on phase I trials of PD-1/PD-L1 inhibitors. Eur. J. Cancer 2018, 89, 56–63. [Google Scholar] [CrossRef] [PubMed]
  167. Xiong, Q.; Huang, Z.; Xin, L.; Qin, B.; Zhao, X.; Zhang, J.; Shi, W.; Yang, B.; Zhang, G.; Hu, Y. Post-treatment neutrophil-to-lymphocyte ratio (NLR) predicts response to anti-PD-1/PD-L1 antibody in SCLC patients at early phase. Cancer Immunol. Immunother. 2020, 70, 713–720. [Google Scholar] [CrossRef] [PubMed]
  168. Shi, Y.; Zhang, J.; Mao, Z.; Jiang, H.; Liu, W.; Shi, H.; Ji, R.; Xu, W.; Qian, H.; Zhang, X. Extracellular Vesicles from Gastric Cancer Cells Induce PD-L1 Expression on Neutrophils to Suppress T-Cell Immunity. Front. Oncol. 2020, 10, 629. [Google Scholar] [CrossRef] [PubMed]
  169. Wang, T.-T.; Zhao, Y.-L.; Peng, L.-S.; Chen, N.; Chen, W.; Lv, Y.-P.; Mao, F.-Y.; Zhang, J.-Y.; Cheng, P.; Teng, Y.-S.; et al. Tumour-activated neutrophils in gastric cancer foster immune suppression and disease progression through GM-CSF-PD-L1 pathway. Gut 2017, 66, 1900–1911. [Google Scholar] [CrossRef] [Green Version]
  170. He, G.; Zhang, H.; Zhou, J.; Wang, B.; Chen, Y.; Kong, Y.; Xie, X.; Wang, X.; Fei, R.; Wei, L. Peritumoural neutrophils negatively regulate adaptive immunity via the PD-L1/PD-1 signalling pathway in hepatocellular carcinoma. J. Exp. Clin. Cancer Res. 2015, 34, 141. [Google Scholar] [CrossRef] [Green Version]
  171. Banna, G.L.; Signorelli, D.; Metro, G.; Galetta, D.; De Toma, A.; Cantale, O.; Banini, M.; Friedlaender, A.; Pizzutillo, P.; Garassino, M.C.; et al. Neutrophil-to-lymphocyte ratio in combination with PD-L1 or lactate dehydrogenase as biomarkers for high PD-L1 non-small cell lung cancer treated with first-line pembrolizumab. Transl. Lung Cancer Res. 2020, 9, 1533–1542. [Google Scholar] [CrossRef]
  172. Kfoury, Y.; Baryawno, N.; Severe, N.; Mei, S.; Gustafsson, K.; Hirz, T.; Brouse, T.; Scadden, E.W.; Igolkina, A.A.; Kokkaliaris, K.; et al. Human prostate cancer bone metastases have an actionable immunosuppressive microenvironment. Cancer Cell 2021, 39, 1464–1478.e8. [Google Scholar] [CrossRef] [PubMed]
  173. DeNardo, D.G.; Ruffell, B. Macrophages as regulators of tumour immunity and immunotherapy. Nat. Rev. Immunol. 2019, 19, 369–382. [Google Scholar] [CrossRef] [PubMed]
  174. Zhang, Q.W.; Liu, L.; Gong, C.-Y.; Shi, H.-S.; Zeng, Y.-H.; Wang, X.-Z.; Zhao, Y.-W.; Wei, Y.-Q. Prognostic Significance of Tumor-Associated Macrophages in Solid Tumor: A Meta-Analysis of the Literature. PLoS ONE 2012, 7, e50946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Mahoney, K.M.; Freeman, G.J.; McDermott, D.F. The Next Immune-Checkpoint Inhibitors: PD-1/PD-L1 Blockade in Melanoma. Clin. Ther. 2015, 37, 764–782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Cai, H.; Zhang, Y.; Wang, J.; Gu, J. Defects in Macrophage Reprogramming in Cancer Therapy: The Negative Impact of PD-L1/PD-1. Front. Immunol. 2021, 12, 690869. [Google Scholar] [CrossRef] [PubMed]
  177. Goldman, N.; Lomakova, Y.D.; Londregan, J.; Bucknum, A.; DePierri, K.; Somerville, J.; Riggs, J.E. High macrophage PD-L1 expression not responsible for T cell suppression. Cell. Immunol. 2018, 324, 50–58. [Google Scholar] [CrossRef]
  178. 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. 2019, 26, 970–977. [Google Scholar] [CrossRef] [Green Version]
  179. Mccord, R.; Bolen, C.R.; Koeppen, H.; Kadel, I.E.E.; Oestergaard, M.Z.; Nielsen, T.; Sehn, L.H.; Venstrom, J.M. PD-L1 and tumor-associated macrophages in de novo DLBCL. Blood Adv. 2019, 3, 531–540. [Google Scholar] [CrossRef]
  180. Schalper, K.A.; Carvajal-Hausdorf, D.; McLaughlin, J.; Velcheti, V.; Chen, L.; Sanmamed, M.; Herbst, R.S.; Rimm, D.L. Clinical significance of PD-L1 protein expression on tumor-associated macrophages in lung cancer. J. Immunother. Cancer 2015, 3, P415. [Google Scholar] [CrossRef] [Green Version]
  181. Kuang, D.-M.; Zhao, Q.; Peng, C.; Xu, J.; Zhang, J.-P.; Wu, C.; Zheng, L. Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J. Exp. Med. 2009, 206, 1327–1337. [Google Scholar] [CrossRef]
  182. Wang, J.; Browne, L.; Slapetova, I.; Shang, F.; Lee, K.; Lynch, J.; Beretov, J.; Whan, R.; Graham, P.H.; Millar, E.K.A. Multiplexed immunofluorescence identifies high stromal CD68+PD-L1+ macrophages as a predictor of improved survival in triple negative breast cancer. Sci. Rep. 2021, 11, 21608. [Google Scholar] [CrossRef] [PubMed]
  183. Simonds, E.F.; Lu, E.D.; Badillo, O.; Karimi, S.; Liu, E.V.; Tamaki, W.; Rancan, C.; Downey, K.M.; Stultz, J.; Sinha, M.; et al. Deep immune profiling reveals targetable mechanisms of immune evasion in immune checkpoint inhibitor-refractory glioblastoma. J. Immunother. Cancer 2021, 9, e002181. [Google Scholar] [CrossRef] [PubMed]
  184. Vilain, R.E.; Menzies, A.; 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] [PubMed] [Green Version]
  185. Toki, M.I.; Merritt, C.R.; Wong, P.F.; Smithy, J.W.; Kluger, H.M.; Syrigos, K.N.; Ong, G.T.; Warren, S.E.; Beechem, J.M.; Rimm, D.L. High-Plex Predictive Marker Discovery for Melanoma Immunotherapy–Treated Patients Using Digital Spatial Profiling. Clin. Cancer Res. 2019, 25, 5503–5512. [Google Scholar] [CrossRef]
  186. Furuse, M.; Kuwabara, H.; Ikeda, N.; Hattori, Y.; Ichikawa, T.; Kagawa, N.; Kikuta, K.; Tamai, S.; Nakada, M.; Wakabayashi, T.; et al. PD-L1 and PD-L2 expression in the tumor microenvironment including peritumoral tissue in primary central nervous system lymphoma. BMC Cancer 2020, 20, 277. [Google Scholar] [CrossRef]
  187. Goswami, S.; Basu, S.; Sharma, P. A potential biomarker for anti-PD-1 immunotherapy. Nat. Med. 2018, 24, 123–124. [Google Scholar] [CrossRef]
  188. Krieg, C.; Nowicka, M.; Guglietta, S.; Schindler, S.; Hartmann, F.J.; Weber, L.M.; Dummer, R.; Robinson, M.D.; Levesque, M.P.; Becher, B. High-dimensional single-cell analysis predicts response to anti-PD-1 immunotherapy. Nat. Med. 2018, 24, 144–153. [Google Scholar] [CrossRef]
  189. Ando, K.; Hamada, K.; Shida, M.; Ohkuma, R.; Kubota, Y.; Horiike, A.; Matsui, H.; Ishiguro, T.; Hirasawa, Y.; Ariizumi, H.; et al. A high number of PD-L1+ CD14+ monocytes in peripheral blood is correlated with shorter survival in patients receiving immune checkpoint inhibitors. Cancer Immunol. Immunother. 2021, 70, 337–348. [Google Scholar] [CrossRef]
  190. Riemann, D.; Schütte, W.; Turzer, S.; Seliger, B.; Möller, M. High PD-L1/CD274 Expression of Monocytes and Blood Dendritic Cells Is a Risk Factor in Lung Cancer Patients Undergoing Treatment with PD1 Inhibitor Therapy. Cancers 2020, 12, 2966. [Google Scholar] [CrossRef]
  191. de Coaña, Y.P.; Wolodarski, M.; Àvila, I.V.D.H.; Nakajima, T.; Rentouli, S.; Lundqvist, A.; Masucci, G.; Hansson, J.; Kiessling, R. PD-1 checkpoint blockade in advanced melanoma patients: NK cells, monocytic subsets and host PD-L1 expression as predictive biomarker candidates. OncoImmunology 2020, 9, 1786888. [Google Scholar] [CrossRef]
  192. Grover, A.; Sanseviero, E.; Timosenko, E.; Gabrilovich, D.I. Myeloid-Derived Suppressor Cells: A Propitious Road to Clinic. Cancer Discov. 2021, 11, 2693–2706. [Google Scholar] [CrossRef] [PubMed]
  193. Davis, R.; Moore, E.C.; Clavijo, P.E.; Friedman, J.; Cash, H.; Chen, Z.; Silvin, C.; Van Waes, C.; Allen, C. Anti-PD-L1 Efficacy Can Be Enhanced by Inhibition of Myeloid-Derived Suppressor Cells with a Selective Inhibitor of PI3Kδ/γ. Cancer Res. 2017, 77, 2607–2619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Noman, M.Z.; Desantis, G.; Janji, B.; Hasmim, M.; Karray, S.; Dessen, P.; Bronte, V.; Chouaib, S. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J. Exp. Med. 2014, 211, 781–790. [Google Scholar] [CrossRef] [PubMed]
  195. Limagne, E.; Euvrard, R.; Thibaudin, M.; Rébé, C.; Derangère, V.; Chevriaux, A.; Boidot, R.; Végran, F.; Bonnefoy, N.; Vincent, J.; et al. Accumulation of MDSC and Th17 Cells in Patients with Metastatic Colorectal Cancer Predicts the Efficacy of a FOLFOX–Bevacizumab Drug Treatment Regimen. Cancer Res. 2016, 76, 5241–5252. [Google Scholar] [CrossRef] [Green Version]
  196. Wang, J.-C.; Chen, C.; Gotlieb, V.; Nalghranyan, S.; Wong, C.; Yeo, I. Elevated Levels of PD-L1 on MDSCs in Patients with Ph(-) Myeloproliferative Neoplasm. Blood 2021, 138, 4591. [Google Scholar] [CrossRef]
  197. Cassetta, L.; Bruderek, K.; Skrzeczynska-Moncznik, J.; Osiecka, O.; Hu, X.; Rundgren, I.M.; Lin, A.; Santegoets, K.; Horzum, U.; Godinho-Santos, A.; et al. Differential expansion of circulating human MDSC subsets in patients with cancer, infection and inflammation. J. Immunother. Cancer 2020, 8, e001223. [Google Scholar] [CrossRef]
  198. Passaro, A.; Mancuso, P.; Gandini, S.; Spitaleri, G.; Labanca, V.; Guerini-Rocco, E.; Barberis, M.; Catania, C.; Del Signore, E.; de Marinis, F.; et al. Gr-MDSC-linked asset as a potential immune biomarker in pretreated NSCLC receiving nivolumab as second-line therapy. Clin. Transl. Oncol. 2020, 22, 603–611. [Google Scholar] [CrossRef]
  199. Sakuishi, K.; Jayaraman, P.; Behar, S.M.; Anderson, A.C.; Kuchroo, V.K. Emerging Tim-3 functions in antimicrobial and tumor immunity. Trends Immunol. 2011, 32, 345–349. [Google Scholar] [CrossRef] [Green Version]
  200. Cabeza-Cabrerizo, M.; Cardoso, A.; Minutti, C.M.; Pereira da Costa, M.; Reis e Sousa, C. Dendritic cells revisited. Annu. Rev. Immunol. 2021, 39, 131–166. [Google Scholar] [CrossRef]
  201. Marciscano, A.E.; Anandasabapathy, N. The role of dendritic cells in cancer and anti-tumor immunity. Semin. Immunol. 2021, 52, 101481. [Google Scholar] [CrossRef]
  202. Bol, K.F.; Schreibelt, G.; Rabold, K.; Wculek, S.K.; Schwarze, J.K.; Dzionek, A.; Teijeira, A.; Kandalaft, L.E.; Romero, P.; Coukos, G.; et al. The clinical application of cancer immunotherapy based on naturally circulating dendritic cells. J. Immunother. Cancer 2019, 7, 109. [Google Scholar] [CrossRef] [PubMed]
  203. Wculek, S.K.; Cueto, F.J.; Mujal, A.M.; Melero, I.; Krummel, M.F.; Sancho, D. Dendritic cells in cancer immunology and immunotherapy. Nat. Rev. Immunol. 2020, 20, 7–24. [Google Scholar] [CrossRef] [PubMed]
  204. Gardner, A.; Ruffell, B. Dendritic Cells and Cancer Immunity. Trends Immunol. 2016, 37, 855–865. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Yin, X.; Chen, S.; Eisenbarth, S.C. Dendritic Cell Regulation of T Helper Cells. Annu. Rev. Immunol. 2021, 39, 759–790. [Google Scholar] [CrossRef] [PubMed]
  206. Reizis, B. Plasmacytoid Dendritic Cells: Development, Regulation, and Function. Immunity 2019, 50, 37–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Fu, C.; Jiang, A. Dendritic Cells and CD8 T Cell Immunity in Tumor Microenvironment. Front. Immunol. 2018, 9, 3059. [Google Scholar] [CrossRef] [Green Version]
  208. Lin, J.H.; Huffman, A.P.; Wattenberg, M.M.; Walter, D.; Carpenter, E.L.; Feldser, D.M.; Beatty, G.L.; Furth, E.E.; Vonderheide, R.H. Type 1 conventional dendritic cells are systemically dysregulated early in pancreatic carcinogenesis. J. Exp. Med. 2020, 217, e20190673. [Google Scholar] [CrossRef]
  209. Palucka, K.; Banchereau, J. Cancer immunotherapy via dendritic cells. Nat. Cancer 2012, 12, 265–277. [Google Scholar] [CrossRef]
  210. Lee, Y.S.; O’Brien, L.J.; Walpole, C.M.; Pearson, F.E.; Leal-Rojas, I.M.; Masterman, K.-A.; Atkinson, V.; Barbour, A.; Radford, K.J. Human CD141+ dendritic cells (cDC1) are impaired in patients with advanced melanoma but can be targeted to enhance anti-PD-1 in a humanized mouse model. J. Immunother. Cancer 2021, 9, e001963. [Google Scholar] [CrossRef]
  211. Melaiu, O.; Chierici, M.; Lucarini, V.; Jurman, G.; Conti, L.A.; De Vito, R.; Boldrini, R.; Cifaldi, L.; Castellano, A.; Furlanello, C.; et al. Cellular and gene signatures of tumor-infiltrating dendritic cells and natural-killer cells predict prognosis of neuroblastoma. Nat. Commun. 2020, 11, 5992. [Google Scholar] [CrossRef]
  212. Oshi, M.; Newman, S.; Tokumaru, Y.; Yan, L.; Matsuyama, R.; Kalinski, P.; Endo, I.; Takabe, K. Plasmacytoid Dendritic Cell (pDC) Infiltration Correlate with Tumor Infiltrating Lymphocytes, Cancer Immunity, and Better Survival in Triple Negative Breast Cancer (TNBC) More Strongly than Conventional Dendritic Cell (cDC). Cancers 2020, 12, 3342. [Google Scholar] [CrossRef] [PubMed]
  213. Mastelic-Gavillet, B.; Sarivalasis, A.; Lozano, L.E.; Wyss, T.; Inoges, S.; de Vries, I.J.M.; Dartiguenave, F.; Jichlinski, P.; Derrè, L.; Coukos, G.; et al. Quantitative and qualitative impairments in dendritic cell subsets of patients with ovarian or prostate cancer. Eur. J. Cancer 2020, 135, 173–182. [Google Scholar] [CrossRef] [PubMed]
  214. Mayoux, M.; Roller, A.; Pulko, V.; Sammicheli, S.; Chen, S.; Sum, E.; Jost, C.; Fransen, M.F.; Buser, R.B.; Kowanetz, M.; et al. Dendritic cells dictate responses to PD-L1 blockade cancer immunotherapy. Sci. Transl. Med. 2020, 12, eaav7431. [Google Scholar] [CrossRef] [PubMed]
  215. Oh, S.A.; Wu, D.-C.; Cheung, J.; Navarro, A.; Xiong, H.; Cubas, R.; Totpal, K.; Chiu, H.; Wu, Y.; Comps-Agrar, L.; et al. PD-L1 expression by dendritic cells is a key regulator of T-cell immunity in cancer. Nat. Cancer 2020, 1, 681–691. [Google Scholar] [CrossRef] [PubMed]
  216. Peng, Q.; Qiu, X.; Zhang, Z.; Zhang, S.; Zhang, Y.; Liang, Y.; Guo, J.; Peng, H.; Chen, M.; Fu, Y.-X.; et al. PD-L1 on dendritic cells attenuates T cell activation and regulates response to immune checkpoint blockade. Nat. Commun. 2020, 11, 4835. [Google Scholar] [CrossRef]
  217. Liu, L.; Chen, J.; Bae, J.; Li, H.; Sun, Z.; Moore, C.; Hsu, E.; Han, C.; Qiao, J.; Fu, Y.-X. Rejuvenation of tumour-specific T cells through bispecific antibodies targeting PD-L1 on dendritic cells. Nat. Biomed. Eng. 2021, 5, 1261–1273. [Google Scholar] [CrossRef]
  218. Lucas, E.D.; Schafer, J.B.; Matsuda, J.; Kraus, M.; Burchill, M.A.; Tamburini, B.A.J. PD-L1 Reverse Signaling in Dermal Dendritic Cells Promotes Dendritic Cell Migration Required for Skin Immunity. Cell Rep. 2020, 33, 108258. [Google Scholar] [CrossRef]
  219. Miller, T.J.; Anyaegbu, C.C.; Lee-Pullen, T.F.; Spalding, L.J.; Platell, C.F.; McCoy, M.J. PD-L1+ dendritic cells in the tumor microenvironment correlate with good prognosis and CD8+ T cell infiltration in colon cancer. Cancer Sci. 2020, 112, 1173–1183. [Google Scholar] [CrossRef]
  220. 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]
  221. Dammeijer, F.; van Gulijk, M.; Mulder, E.E.; Lukkes, M.; Klaase, L.; Bosch, T.V.D.; van Nimwegen, M.; Lau, S.P.; Latupeirissa, K.; Schetters, S.; et al. The PD-1/PD-L1-Checkpoint Restrains T cell Immunity in Tumor-Draining Lymph Nodes. Cancer Cell 2020, 38, 685–700.e8. [Google Scholar] [CrossRef]
  222. Lin, H.; Wei, S.; Hurt, E.M.; Green, M.D.; Zhao, L.; Vatan, L.; Szeliga, W.; Herbst, R.; Harms, P.W.; Fecher, L.A.; et al. Host expression of PD-L1 determines efficacy of PD-L1 pathway blockade–mediated tumor regression. J. Clin. Investig. 2018, 128, 805–815. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. El Bairi, K.; Haynes, H.R.; Blackley, E.; Fineberg, S.; Shear, J.; Turner, S.; de Freitas, J.R.; Sur, D.; Amendola, L.C.; Gharib, M.; et al. The tale of TILs in breast cancer: A report from The International Immuno-Oncology Biomarker Working Group. npj Breast Cancer 2021, 7, 150. [Google Scholar] [CrossRef] [PubMed]
  224. Gonzalez-Ericsson, P.I.; Stovgaard, E.S.; Sua, L.F.; Reisenbichler, E.; Kos, Z.; Carter, J.M.; Michiels, S.; Le Quesne, J.; Nielsen, T.O.; Lænkholm, A.V.; et al. The path to a better biomarker: Application of a risk management framework for the implementation of PD-L1 and TILs as immuno-oncology biomarkers in breast cancer clinical trials and daily practice. J. Pathol. 2020, 250, 667–684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Miyake, M.; Hori, S.; Owari, T.; Oda, Y.; Tatsumi, Y.; Nakai, Y.; Fujii, T.; Fujimoto, K. Clinical Impact of Tumor-Infiltrating Lymphocytes and PD-L1-Positive Cells as Prognostic and Predictive Biomarkers in Urological Malignancies and Retroperitoneal Sarcoma. Cancers 2020, 12, 3153. [Google Scholar] [CrossRef] [PubMed]
  226. Uryvaev, A.; Passhak, M.; Hershkovits, D.; Sabo, E.; Bar-Sela, G. The role of tumor-infiltrating lymphocytes (TILs) as a predictive biomarker of response to anti-PD1 therapy in patients with metastatic non-small cell lung cancer or metastatic melanoma. Med. Oncol. 2018, 35, 25. [Google Scholar] [CrossRef]
  227. Hadrup, S.; Donia, M.; Straten, P.T. Effector CD4 and CD8 T Cells and Their Role in the Tumor Microenvironment. Cancer Microenviron. 2013, 6, 123–133. [Google Scholar] [CrossRef] [Green Version]
  228. Budimir, N.; Thomas, G.D.; Dolina, J.S.; Salek-Ardakani, S. Reversing T-cell Exhaustion in Cancer: Lessons Learned from PD-1/PD-L1 Immune Checkpoint Blockade. Cancer Immunol. Res. 2021, 10, 146–153. [Google Scholar] [CrossRef]
  229. Van Der Leun, A.M.; Thommen, D.S.; Schumacher, T.N. CD8+ T cell states in human cancer: Insights from single-cell analysis. Nat. Cancer 2020, 20, 218–232. [Google Scholar] [CrossRef]
  230. Gibney, G.T.; Weiner, L.M.; Atkins, M.B. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 2016, 17, e542–e551. [Google Scholar] [CrossRef] [Green Version]
  231. Lee, J.; Lozano-Ruiz, B.; Yang, F.M.; Fan, D.D.; Shen, L.; González-Navajas, J.M. The Multifaceted Role of Th1, Th9, and Th17 Cells in Immune Checkpoint Inhibition Therapy. Front. Immunol. 2021, 12, 625667. [Google Scholar] [CrossRef]
  232. Niogret, J.; Berger, H.; Rebe, C.; Mary, R.; Ballot, E.; Truntzer, C.; Thibaudin, M.; Derangère, V.; Hibos, C.; Hampe, L.; et al. Follicular helper-T cells restore CD8+-dependent antitumor immunity and anti-PD-L1/PD-1 efficacy. J. Immunother. Cancer 2021, 9, e002157. [Google Scholar] [CrossRef] [PubMed]
  233. Miller, B.C.; Sen, D.R.; Al Abosy, R.; 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] [PubMed]
  234. Jiang, P.; Gu, S.; Pan, D.; Fu, J.; Sahu, A.; Hu, X.; Li, Z.; Traugh, N.; Bu, X.; Li, B.; et al. Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nat. Med. 2018, 24, 1550–1558. [Google Scholar] [CrossRef] [PubMed]
  235. Byrne, A.; Savas, P.; Sant, S.; Li, R.; Virassamy, B.; Luen, S.J.; Beavis, P.; Mackay, L.K.; Neeson, P.J.; Loi, S. Tissue-resident memory T cells in breast cancer control and immunotherapy responses. Nat. Rev. Clin. Oncol. 2020, 17, 341–348. [Google Scholar] [CrossRef] [PubMed]
  236. Duchemann, B.; Remon, J.; Naigeon, M.; Mezquita, L.; Ferrara, R.; Cassard, L.; Jouniaux, J.M.; Boselli, L.; Grivel, J.; Auclin, E.; et al. Integrating Circulating Biomarkers in the Immune Checkpoint Inhibitor Treatment in Lung Cancer. Cancers 2020, 12, 3625. [Google Scholar] [CrossRef]
  237. An, H.J.; Chon, H.J.; Kim, C. Peripheral Blood-Based Biomarkers for Immune Checkpoint Inhibitors. Int. J. Mol. Sci. 2021, 22, 9414. [Google Scholar] [CrossRef]
  238. Duchemann, B.; Naigeon, M.; Auclin, E.; Ferrara, R.; Cassard, L.; Jouniaux, J.-M.; Boselli, L.; Grivel, J.; Desnoyer, A.; Danlos, F.-X.; et al. CD8+PD-1+ to CD4+PD-1+ ratio (PERLS) is associated with prognosis of patients with advanced NSCLC treated with PD-(L)1 blockers. J. Immunother. Cancer 2022, 10, e004012. [Google Scholar] [CrossRef]
  239. Lucca, L.E.; Axisa, P.-P.; Lu, B.; Harnett, B.; Jessel, S.; Zhang, L.; Raddassi, K.; Zhang, L.; Olino, K.; Clune, J.; et al. Circulating clonally expanded T cells reflect functions of tumor-infiltrating T cells. J. Exp. Med. 2021, 218, e20200921. [Google Scholar] [CrossRef]
  240. Kang, D.H.; Chung, C.; Sun, P.; Lee, D.H.; Lee, S.-I.; Park, D.; Koh, J.S.; Kim, Y.; Yi, H.-S.; Lee, J.E. Circulating regulatory T cells predict efficacy and atypical responses in lung cancer patients treated with PD-1/PD-L1 inhibitors. Cancer Immunol. Immunother. 2022, 71, 579–588. [Google Scholar] [CrossRef]
  241. De Biasi, S.; Gibellini, L.; Tartaro, D.L.; Puccio, S.; Rabacchi, C.; Mazza, E.M.C.; Brummelman, J.; Williams, B.; Kaihara, K.; Forcato, M.; et al. Circulating mucosal-associated invariant T cells identify patients responding to anti-PD-1 therapy. Nat. Commun. 2021, 12, 1669. [Google Scholar] [CrossRef]
  242. Zuazo, M.; Arasanz, H.; Bocanegra, A.; Chocarro, L.; Vera, R.; Escors, D.; Kagamu, H.; Kochan, G. Systemic CD4 immunity: A powerful clinical biomarker for PD-L1/PD-1 immunotherapy. EMBO Mol. Med. 2020, 12, e12706. [Google Scholar] [CrossRef] [PubMed]
  243. Diskin, B.; Adam, S.; Cassini, M.F.; Sanchez, G.; Liria, M.; Aykut, B.; Buttar, C.; Li, E.; Sundberg, B.; Salas, R.D.; et al. PD-L1 engagement on T cells promotes self-tolerance and suppression of neighboring macrophages and effector T cells in cancer. Nat. Immunol. 2020, 21, 442–454. [Google Scholar] [CrossRef] [PubMed]
  244. Jacquelot, N.; Roberti, M.P.; Enot, D.P.; Rusakiewicz, S.; Ternès, N.; Jegou, S.; Woods, D.M.; Sodré, A.L.; Hansen, M.; Meirow, Y.; et al. Predictors of responses to immune checkpoint blockade in advanced melanoma. Nat. Commun. 2017, 8, 592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Castro, F.; Cardoso, A.P.; Gonçalves, R.M.; Serre, K.; Oliveira, M.J. Interferon-Gamma at the Crossroads of Tumor Immune Surveillance or Evasion. Front. Immunol. 2018, 9, 847. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  246. Ivashkiv, L.B. IFNγ: Signalling, epigenetics and roles in immunity, metabolism, disease and cancer immunotherapy. Nat. Rev. Immunol. 2018, 18, 545–558. [Google Scholar] [CrossRef]
  247. Garcia-Diaz, A.; Shin, D.S.; Moreno, B.H.; Saco, J.; Escuin-Ordinas, H.; Rodriguez, G.A.; Zaretsky, J.M.; Sun, L.; Hugo, W.; Wang, X.; et al. Interferon Receptor Signaling Pathways Regulating PD-L1 and PD-L2 Expression. Cell Rep. 2017, 19, 1189–1201. [Google Scholar] [CrossRef] [Green Version]
  248. Naidus, E.; Bouquet, J.; Oh, D.Y.; Looney, T.J.; Yang, H.; Fong, L.; Standifer, N.E.; Zhang, L. Early changes in the circulating T cells are associated with clinical outcomes after PD-L1 blockade by durvalumab in advanced NSCLC patients. Cancer Immunol. Immunother. 2021, 70, 2095–2102. [Google Scholar] [CrossRef]
  249. Bacot, S.M.; Harper, T.A.; Matthews, R.L.; Fennell, C.J.; Akue, A.; Kukuruga, M.A.; Lee, S.; Wang, T.; Feldman, G.M. Exploring the Potential Use of a PBMC-Based Functional Assay to Identify Predictive Biomarkers for Anti-PD-1 Immunotherapy. Int. J. Mol. Sci. 2020, 21, 9023. [Google Scholar] [CrossRef]
  250. Shen, R.; Postow, M.A.; Adamow, M.; Arora, A.; Hannum, M.; Maher, C.; Wong, P.; Curran, M.A.; Hollmann, T.J.; Jia, L.; et al. LAG-3 expression on peripheral blood cells identifies patients with poorer outcomes after immune checkpoint blockade. Sci. Transl. Med. 2021, 13, eabf5107. [Google Scholar] [CrossRef]
  251. Tawbi, H.A.; Schadendorf, D.; Lipson, E.J.; Ascierto, P.A.; Matamala, L.; Gutiérrez, E.C.; Rutkowski, P.; Gogas, H.J.; Lao, C.D.; De Menezes, J.J.; et al. Relatlimab and Nivolumab versus Nivolumab in Untreated Advanced Melanoma. N. Engl. J. Med. 2022, 386, 24–34. [Google Scholar] [CrossRef]
  252. Esfahani, K.; Elkrief, A.; Calabrese, C.; Lapointe, R.; Hudson, M.; Routy, B.; Miller, W.H., Jr.; Calabrese, L. Moving towards personalized treatments of immune-related adverse events. Nat. Rev. Clin. Oncol. 2020, 17, 504–515. [Google Scholar] [CrossRef] [PubMed]
  253. Cristescu, R.; Mogg, R.; Ayers, M.; Albright, A.; Murphy, E.; Yearley, J.; Sher, X.; Liu, X.Q.; Lu, H.; Nebozhyn, M.; et al. Pan-tumor genomic biomarkers for PD-1 checkpoint blockade–based immunotherapy. Science 2018, 362, eaar3593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Petitprez, F.; de Reyniès, A.; Keung, E.Z.; Chen, T.W.-W.; Sun, C.-M.; Calderaro, J.; Jeng, Y.-M.; Hsiao, L.-P.; Lacroix, L.; Bougoüin, A.; et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature 2020, 577, 556–560. [Google Scholar] [CrossRef] [PubMed]
  255. Cabrita, R.; Lauss, M.; Sanna, A.; Donia, M.; Larsen, M.S.; Mitra, S.; Johansson, I.; Phung, B.; Harbst, K.; Vallon-Christersson, J.; et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature 2020, 577, 561–565. [Google Scholar] [CrossRef] [PubMed]
  256. Romero, D. B cells and TLSs facilitate a response to ICI. Nat. Rev. Clin. Oncol. 2020, 17, 195. [Google Scholar] [CrossRef]
  257. Helmink, B.A.; Reddy, S.M.; Gao, J.; Zhang, S.; Basar, R.; Thakur, R.; Yizhak, K.; Sade-Feldman, M.; Blando, J.; Han, G.; et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature 2020, 577, 549–555. [Google Scholar] [CrossRef] [PubMed]
  258. Patil, N.S.; Nabet, B.Y.; Müller, S.; Koeppen, H.; Zou, W.; Giltnane, J.; Au-Yeung, A.; Srivats, S.; Cheng, J.H.; Takahashi, C.; et al. Intratumoral plasma cells predict outcomes to PD-L1 blockade in non-small cell lung cancer. Cancer Cell 2022, 40, 289–300.e4. [Google Scholar] [CrossRef] [PubMed]
  259. Cózar, B.; Greppi, M.; Carpentier, S.; Narni-Mancinelli, E.; Chiossone, L.; Vivier, E. Tumor-Infiltrating Natural Killer Cells. Cancer Discov. 2021, 11, 34–44. [Google Scholar] [CrossRef]
  260. Hsu, J.; Hodgins, J.J.; Marathe, M.; Nicolai, C.J.; Bourgeois-Daigneault, M.-C.; Trevino, T.N.; Azimi, C.S.; Scheer, A.K.; Randolph, H.E.; Thompson, T.W.; et al. Contribution of NK cells to immunotherapy mediated by PD-1/PD-L1 blockade. J. Clin. Investig. 2018, 128, 4654–4668. [Google Scholar] [CrossRef]
  261. Sun, X.; Zhang, T.; Li, M.; Yin, L.; Xue, J. Immunosuppressive B cells expressing PD-1/PD-L1 in solid tumors: A mini review. QJM Int. J. 2019, hcz162. [Google Scholar] [CrossRef]
  262. Lau, J.; Cheung, J.; Navarro, A.; Lianoglou, S.; Haley, B.; Totpal, K.; Sanders, L.; Koeppen, H.; Caplazi, P.; McBride, J.; et al. Tumour and host cell PD-L1 is required to mediate suppression of anti-tumour immunity in mice. Nat. Commun. 2017, 8, 14572. [Google Scholar] [CrossRef] [PubMed]
  263. Chevrier, S.; Levine, J.H.; Zanotelli, V.R.T.; Silina, K.; Schulz, D.; Bacac, M.; Ries, C.H.; Ailles, L.; Jewett, M.A.S.; Moch, H.; et al. An Immune Atlas of Clear Cell Renal Cell Carcinoma. Cell 2017, 169, 736–749.e18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  264. Kotecha, R.R.; Motzer, R.J.; Voss, M.H. Towards individualized therapy for metastatic renal cell carcinoma. Nat. Rev. Clin. Oncol. 2019, 16, 621–633. [Google Scholar] [CrossRef]
  265. Donahue, R.N.; Lepone, L.M.; Grenga, I.; Jochems, C.; Fantini, M.; Madan, R.A.; Heery, C.R.; Gulley, J.L.; Schlom, J. Analyses of the peripheral immunome following multiple administrations of avelumab, a human IgG1 anti-PD-L1 monoclonal antibody. J. Immunother. Cancer 2017, 5, 20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  266. Yang, J.-M.; Chi, W.-Y.; Liang, J.; Takayanagi, S.; Iglesias, P.A.; Huang, C.-H. Deciphering cell signaling networks with massively multiplexed biosensor barcoding. Cell 2021, 184, 6193–6206.e14. [Google Scholar] [CrossRef]
  267. Boehm, K.M.; Khosravi, P.; Vanguri, R.; Gao, J.; Shah, S.P. Harnessing multimodal data integration to advance precision oncology. Nat. Cancer 2021, 22, 114–126. [Google Scholar] [CrossRef]
  268. Adler-Milstein, J.; Chen, J.H.; Dhaliwal, G. Next-Generation Artificial Intelligence for Diagnosis: From Predicting Diagnostic Labels to “Wayfinding”. JAMA 2021, 326, 2467–2468. [Google Scholar] [CrossRef]
  269. Ciccolini, J.; Benzekry, S.; Barlesi, F. Deciphering the response and resistance to immune-checkpoint inhibitors in lung cancer with artificial intelligence-based analysis: When PIONeeR meets QUANTIC. Br. J. Cancer 2020, 123, 337–338. [Google Scholar] [CrossRef]
  270. Elemento, O.; Leslie, C.; Lundin, J.; Tourassi, G. Artificial intelligence in cancer research, diagnosis and therapy. Nat. Cancer 2021, 21, 747–752. [Google Scholar] [CrossRef]
  271. Ho, D. Artificial intelligence in cancer therapy. Science 2020, 367, 982–983. [Google Scholar] [CrossRef]
  272. Gide, T.N.; Silva, I.P.; Quek, C.; Ahmed, T.; Menzies, A.M.; Carlino, M.S.; Saw, R.P.; Thompson, J.F.; Batten, M.; Long, G.V.; et al. Close proximity of immune and tumor cells underlies response to anti-PD-1 based therapies in metastatic melanoma patients. OncoImmunology 2020, 9, 1659093. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  273. Noguchi, T.; Ward, J.; Gubin, M.M.; Arthur, C.D.; Lee, S.H.; Hundal, J.; Selby, M.J.; Graziano, R.F.; Mardis, E.R.; Korman, A.J.; et al. Temporally Distinct PD-L1 Expression by Tumor and Host Cells Contributes to Immune Escape. Cancer Immunol. Res. 2017, 5, 106–117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  274. Fiori, M.E.; Di Franco, S.; Villanova, L.; Bianca, P.; Stassi, G.; De Maria, R. Cancer-associated fibroblasts as abettors of tumor progression at the crossroads of EMT and therapy resistance. Mol. Cancer 2019, 18, 1189–1201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  275. Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef] [PubMed]
  276. Pegtel, D.M.; Gould, S.J. Exosomes. Annu. Rev. Biochem. 2019, 88, 487–514. [Google Scholar] [CrossRef]
  277. Kugeratski, F.G.; Kalluri, R. Exosomes as mediators of immune regulation and immunotherapy in cancer. FEBS J. 2021, 288, 10–35. [Google Scholar] [CrossRef]
  278. Sharma, P.; Diergaarde, B.; Ferrone, S.; Kirkwood, J.M.; Whiteside, T.L. Melanoma cell-derived exosomes in plasma of melanoma patients suppress functions of immune effector cells. Sci. Rep. 2020, 10, 92. [Google Scholar] [CrossRef]
  279. Shen, M.; Ren, X. New insights into the biological impacts of immune cell-derived exosomes within the tumor environment. Cancer Lett. 2018, 431, 115–122. [Google Scholar] [CrossRef]
  280. Tavasolian, F.; Hosseini, A.Z.; Rashidi, M.; Soudi, S.; Abdollahi, E.; Momtazi-Borojeni, A.A.; Sathyapalan, T.; Sahebkar, A. The Impact of Immune Cell-derived Exosomes on Immune Response Initiation and Immune System Function. Curr. Pharm. Des. 2021, 27, 197–205. [Google Scholar] [CrossRef]
  281. Yan, W.; Jiang, S. Immune Cell-Derived Exosomes in the Cancer-Immunity Cycle. Trends Cancer 2020, 6, 506–517. [Google Scholar] [CrossRef]
  282. Daassi, D.; Mahoney, K.M.; Freeman, G.J. The importance of exosomal PDL1 in tumour immune evasion. Nat. Rev. Immunol. 2020, 20, 209–215. [Google Scholar] [CrossRef] [PubMed]
  283. Li, W.; Li, C.; Zhou, T.; Liu, X.; Liu, X.; Li, X.; Chen, D. Role of exosomal proteins in cancer diagnosis. Mol. Cancer 2017, 16, 145. [Google Scholar] [CrossRef] [PubMed]
  284. Poggio, M.; Hu, T.; Pai, C.-C.; Chu, B.; Belair, C.D.; Chang, A.; Montabana, E.; Lang, U.E.; Fu, Q.; Fong, L.; et al. Suppression of exosomal PD-L1 induces systemic anti-tumor immunity and memory. Cell 2019, 177, 414–427.e413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  285. Chen, G.; Huang, A.C.; Zhang, W.; Zhang, G.; Wu, M.; Xu, W.; Yu, Z.; Yang, J.; Wang, B.; Sun, H.; et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 2018, 560, 382–386. [Google Scholar] [CrossRef] [PubMed]
  286. Liu, J.; Peng, X.; Yang, S.; Li, X.; Huang, M.; Wei, S.; Zhang, S.; He, G.; Zheng, H.; Fan, Q.; et al. Extracellular vesicle PD-L1 in reshaping tumor immune microenvironment: Biological function and potential therapy strategies. Cell Commun. Signal. 2022, 20, 14. [Google Scholar] [CrossRef] [PubMed]
  287. Tang, Y.; Zhang, P.; Wang, Y.; Wang, J.; Su, M.; Wang, Y.; Zhou, L.; Zhou, J.; Xiong, W.; Zeng, Z.; et al. The Biogenesis, Biology, and Clinical Significance of Exosomal PD-L1 in Cancer. Front. Immunol. 2020, 11, 604. [Google Scholar] [CrossRef]
  288. Cordonnier, M.; Nardin, C.; Chanteloup, G.; Derangere, V.; Algros, M.; Arnould, L.; Garrido, C.; Aubin, F.; Gobbo, J. Tracking the evolution of circulating exosomal-PD-L1 to monitor melanoma patients. J. Extracell. Vesicles 2020, 9, 1710899. [Google Scholar] [CrossRef] [Green Version]
  289. Yu, W.; Hurley, J.; Roberts, D.; Chakrabortty, S.; Enderle, D.; Noerholm, M.; Breakefield, X.; Skog, J. Exosome-based liquid biopsies in cancer: Opportunities and challenges. Ann. Oncol. 2021, 32, 466–477. [Google Scholar] [CrossRef]
  290. Zhang, C.; Fan, Y.; Che, X.; Zhang, M.; Li, Z.; Li, C.; Wang, S.; Wen, T.; Hou, K.; Shao, X.; et al. Anti-PD-1 Therapy Response Predicted by the Combination of Exosomal PD-L1 and CD28. Front. Oncol. 2020, 10, 760. [Google Scholar] [CrossRef]
  291. Morrissey, S.M.; Yan, J. Exosomal PD-L1: Roles in Tumor Progression and Immunotherapy. Trends Cancer 2020, 6, 550–558. [Google Scholar] [CrossRef]
  292. Yang, Q.; Chen, M.; Gu, J.; Niu, K.; Zhao, X.; Zheng, L.; Xu, Z.; Yu, Y.; Li, F.; Meng, L.; et al. Novel Biomarkers of Dynamic Blood PD-L1 Expression for Immune Checkpoint Inhibitors in Advanced Non-Small-Cell Lung Cancer Patients. Front. Immunol. 2021, 12, 665133. [Google Scholar] [CrossRef] [PubMed]
  293. Ludwig, N.; Hong, C.; Ludwig, S.; Azambuja, J.H.; Sharma, P.; Theodoraki, M.; Whiteside, T.L. Isolation and Analysis of Tumor-Derived Exosomes. Curr. Protoc. Immunol. 2019, 127, e91. [Google Scholar] [CrossRef] [PubMed]
  294. Mohammadi, M.; Zargartalebi, H.; Salahandish, R.; Aburashed, R.; Yong, K.W.; Sanati-Nezhad, A. Emerging technologies and commercial products in exosome-based cancer diagnosis and prognosis. Biosens. Bioelectron. 2021, 183, 113176. [Google Scholar] [CrossRef] [PubMed]
  295. Chen, Y.; Zhu, Q.; Cheng, L.; Wang, Y.; Li, M.; Yang, Q.; Hu, L.; Lou, D.; Li, J.; Dong, X.; et al. Exosome detection via the ultrafast-isolation system: EXODUS. Nat. Methods 2021, 18, 212–218. [Google Scholar] [CrossRef]
  296. Zhang, P.; Zhou, X.; He, M.; Shang, Y.; Tetlow, A.L.; Godwin, A.K.; Zeng, Y. Ultrasensitive detection of circulating exosomes with a 3D-nanopatterned microfluidic chip. Nat. Biomed. Eng. 2019, 3, 438–451. [Google Scholar] [CrossRef]
  297. Li, C.; Li, C.; Zhi, C.; Liang, W.; Wang, X.; Chen, X.; Lv, T.; Shen, Q.; Song, Y.; Lin, D.; et al. Clinical significance of PD-L1 expression in serum-derived exosomes in NSCLC patients. J. Transl. Med. 2019, 17, 355. [Google Scholar] [CrossRef] [Green Version]
  298. Gong, B.; Kiyotani, K.; Sakata, S.; Nagano, S.; Kumehara, S.; Baba, S.; Besse, B.; Yanagitani, N.; Friboulet, L.; Nishio, M.; et al. Secreted PD-L1 variants mediate resistance to PD-L1 blockade therapy in non–small cell lung cancer. J. Exp. Med. 2019, 216, 982–1000. [Google Scholar] [CrossRef] [Green Version]
  299. Wan, J.C.M.; Heider, K.; Gale, D.; Murphy, S.; Fisher, E.; Mouliere, F.; Ruiz-Valdepenas, A.; Santonja, A.; Morris, J.; Chandrananda, D.; et al. ctDNA monitoring using patient-specific sequencing and integration of variant reads. Sci. Transl. Med. 2020, 12, eaaz8084. [Google Scholar] [CrossRef]
  300. Ishiba, T.; Hoffmann, A.-C.; Usher, J.; Elshimali, Y.; Sturdevant, T.; Dang, M.; Jaimes, Y.; Tyagi, R.; Gonzales, R.; Grino, M.; et al. Frequencies and expression levels of programmed death ligand 1 (PD-L1) in circulating tumor RNA (ctRNA) in various cancer types. Biochem. Biophys. Res. Commun. 2018, 500, 621–625. [Google Scholar] [CrossRef]
  301. Finkelmeier, F.; Canli, Ö.; Tal, A.; Pleli, T.; Trojan, J.; Schmidt, M.; Kronenberger, B.; Zeuzem, S.; Piiper, A.; Greten, F.R. High levels of the soluble programmed death-ligand (sPD-L1) identify hepatocellular carcinoma patients with a poor prognosis. Eur. J. Cancer 2016, 59, 152–159. [Google Scholar] [CrossRef]
  302. Chatterjee, J.; Dai, W.; Aziz, N.H.A.; Teo, P.Y.; Wahba, J.; Phelps, D.L.; Maine, C.J.; Whilding, L.M.; Dina, R.; Trevisan, G.; et al. Clinical Use of Programmed Cell Death-1 and Its Ligand Expression as Discriminatory and Predictive Markers in Ovarian Cancer. Clin. Cancer Res. 2017, 23, 3453–3460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  303. Chang, B.; Huang, T.; Wei, H.; Shen, L.; Zhu, D.; He, W.; Chen, Q.; Zhang, H.; Li, Y.; Huang, R.; et al. The correlation and prognostic value of serum levels of soluble programmed death protein 1 (sPD-1) and soluble programmed death-ligand 1 (sPD-L1) in patients with hepatocellular carcinoma. Cancer Immunol. Immunother. 2019, 68, 353–363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  304. Larrinaga, G.; Solano-Iturri, J.D.; Errarte, P.; Unda, M.; Loizaga-Iriarte, A.; Pérez-Fernández, A.; Echevarría, E.; Asumendi, A.; Manini, C.; Angulo, J.C.; et al. Soluble PD-L1 Is an Independent Prognostic Factor in Clear Cell Renal Cell Carcinoma. Cancers 2021, 13, 667. [Google Scholar] [CrossRef] [PubMed]
  305. Oh, S.Y.; Kim, S.; Keam, B.; Kim, T.M.; Kim, D.-W.; Heo, D.S. Soluble PD-L1 is a predictive and prognostic biomarker in advanced cancer patients who receive immune checkpoint blockade treatment. Sci. Rep. 2021, 11, 19712. [Google Scholar] [CrossRef] [PubMed]
  306. Okuma, Y.; Wakui, H.; Utsumi, H.; Sagawa, Y.; Hosomi, Y.; Kuwano, K.; Homma, S. Soluble Programmed Cell Death Ligand 1 as a Novel Biomarker for Nivolumab Therapy for Non-Small-cell Lung Cancer. Clin. Lung Cancer 2018, 19, 410–417.e1. [Google Scholar] [CrossRef] [PubMed]
  307. Cheng, Y.; Wang, C.; Wang, Y.; Dai, L. Soluble PD-L1 as a predictive biomarker in lung cancer: A systematic review and meta-analysis. Futur. Oncol. 2022, 18, 261–273. [Google Scholar] [CrossRef]
  308. Incorvaia, L.; Fanale, D.; Badalamenti, G.; Porta, C.; Olive, D.; De Luca, I.; Brando, C.; Rizzo, M.; Messina, C.; Rediti, M.; et al. Baseline Plasma Levels of Soluble PD-1, PD-L1, and BTN3A1 Predict Response to Nivolumab Treatment in Patients with Metastatic Renal Cell Carcinoma: A Step Toward a Biomarker for Therapeutic Decisions. OncoImmunology 2020, 9, 1832348. [Google Scholar] [CrossRef]
  309. Mahoney, K.M.; Ross-Macdonald, P.; Yuan, L.; Song, L.; Veras, E.; Wind-Rotolo, M.; McDermott, D.F.; Hodi, F.S.; Choueiri, T.K.; Freeman, G.J. Soluble PD-L1 as an early marker of progressive disease on nivolumab. J. Immunother. Cancer 2022, 10, e003527. [Google Scholar] [CrossRef]
  310. Costantini, A.; Julie, C.; Dumenil, C.; Hélias-Rodzewicz, Z.; Tisserand, J.; Dumoulin, J.; Giraud, V.; Labrune, S.; Chinet, T.; Emile, J.-F.; et al. Predictive role of plasmatic biomarkers in advanced non-small cell lung cancer treated by nivolumab. OncoImmunology 2018, 7, e1452581. [Google Scholar] [CrossRef] [Green Version]
  311. Castello, A.; Rossi, S.; Toschi, L.; Mansi, L.; Lopci, E. Soluble PD-L1 in NSCLC Patients Treated with Checkpoint Inhibitors and Its Correlation with Metabolic Parameters. Cancers 2020, 12, 1373. [Google Scholar] [CrossRef]
  312. Mallardo, D.; Vitale, M.G.; Giannarelli, D.; Trillò, G.; Esposito, A.; Capone, M.; Isgrò, M.A.; Madonna, G.; D’Angelo, G.; Festino, L.; et al. 24 Nivolumab serum concentration in metastatic melanoma patients could be related to anti-tumor activity gene and outcome. J. Immunother. Cancer 2021, 9, A27–A28. [Google Scholar] [CrossRef]
  313. Basak, E.A.; Koolen, S.; Hurkmans, D.P.; Schreurs, M.W.; Bins, S.; de Hoop, E.O.; Wijkhuijs, A.J.; Besten, I.D.; Sleijfer, S.; Debets, R.; et al. Correlation between nivolumab exposure and treatment outcomes in non–small-cell lung cancer. Eur. J. Cancer 2019, 109, 12–20. [Google Scholar] [CrossRef] [PubMed]
  314. Meza, L.; Salgia, N.J.; Patel, K.C.; Pal, S.K. Learning from BISCAY: The future of biomarker-based trial design in bladder cancer. Cancer Cell 2021, 39, 910–912. [Google Scholar] [CrossRef] [PubMed]
  315. Saad, E.D.; Paoletti, X.; Burzykowski, T.; Buyse, M. Precision medicine needs randomized clinical trials. Nat. Rev. Clin. Oncol. 2017, 14, 317–323. [Google Scholar] [CrossRef]
  316. Hu, C.; Dignam, J.J. Biomarker-Driven Oncology Clinical Trials: Key Design Elements, Types, Features, and Practical Considerations. JCO Precis. Oncol. 2019, 3, 1–12. [Google Scholar] [CrossRef] [PubMed]
  317. Redman, M.W.; Papadimitrakopoulou, V.A.; Minichiello, K.; Hirsch, F.R.; Mack, P.C.; Schwartz, L.H.; Vokes, E.; Ramalingam, S.; Leighl, N.; Bradley, J.; et al. Biomarker-driven therapies for previously treated squamous non-small-cell lung cancer (Lung-MAP SWOG S1400): A biomarker-driven master protocol. Lancet Oncol. 2020, 21, 1589–1601. [Google Scholar] [CrossRef]
  318. Antoniou, M.; Kolamunnage-Dona, R.; Wason, J.; Bathia, R.; Billingham, C.; Bliss, J.; Brown, L.; Gillman, A.; Paul, J.; Jorgensen, A. Biomarker-guided trials: Challenges in practice. Contemp. Clin. Trials Commun. 2019, 16, 100493. [Google Scholar] [CrossRef]
  319. Jiang, C.Y.; Niu, Z.; Green, M.D.; Zhao, L.; Raupp, S.; Pannecouk, B.; Brenner, D.E.; Nagrath, S.; Ramnath, N. It’s not ‘just a tube of blood’: Principles of protocol development, sample collection, staffing and budget considerations for blood-based biomarkers in immunotherapy studies. J. Immunother. Cancer 2021, 9, e03212. [Google Scholar] [CrossRef]
  320. McDermott, J.E.; Wang, J.; Mitchell, H.; Webb-Robertson, B.-J.; Hafen, R.; Ramey, J.; Rodland, K.D. Challenges in biomarker discovery: Combining expert insights with statistical analysis of complex omics data. Expert Opin. Med. Diagn. 2013, 7, 37–51. [Google Scholar] [CrossRef] [Green Version]
  321. Simon, R. Review of Statistical Methods for Biomarker-Driven Clinical Trials. JCO Precis. Oncol. 2019, 3, 1–9. [Google Scholar] [CrossRef]
  322. Goswami, S.; Sharma, P. Genetic biomarker for cancer immunotherapy. Science 2017, 357, 358. [Google Scholar] [CrossRef] [PubMed]
  323. Patterson, L.F.S.; Vardhana, S.A. Metabolic regulation of the cancer-immunity cycle. Trends Immunol. 2021, 42, 975–993. [Google Scholar] [CrossRef] [PubMed]
  324. Villanueva, L.; Alvarez_Errico, D.; Esteller, M. The Contribution of Epigenetics to Cancer Immunotherapy. Trends Immunol. 2020, 41, 676–691. [Google Scholar] [CrossRef] [PubMed]
  325. Powles, T.; Carroll, D.; Chowdhury, S.; Gravis, G.; Joly, F.; Carles, J.; Fléchon, A.; Maroto, P.; Petrylak, D.; Rolland, F.; et al. An adaptive, biomarker-directed platform study of durvalumab in combination with targeted therapies in advanced urothelial cancer. Nat. Med. 2021, 27, 793–801. [Google Scholar] [CrossRef]
  326. Lam, K.C.; Goldszmid, R.S. Can gut microbes predict efficacy and toxicity of combined immune checkpoint blockade? Cancer Cell 2021, 39, 1314–1316. [Google Scholar] [CrossRef]
  327. Lennon, A.M.; Buchanan, A.H.; Kinde, I.; Warren, A.; Honushefsky, A.; Cohain, A.T.; Ledbetter, D.H.; Sanfilippo, F.; Sheridan, K.; Rosica, D.; et al. Feasibility of blood testing combined with PET-CT to screen for cancer and guide intervention. Science 2020, 369, eabb9601. [Google Scholar] [CrossRef]
  328. Kaira, K.; Kuji, I.; Kagamu, H. Value of 18F-FDG-PET to predict PD-L1 expression and outcomes of PD-1 inhibition therapy in human cancers. Cancer Imaging 2021, 21, 11. [Google Scholar] [CrossRef]
  329. Rakhshandehroo, T.; Smith, B.R.; Glockner, H.J.; Rashidian, M.; Pandit-Taskar, N. Molecular Immune Targeted Imaging of Tumor Microenvironment. Nanotheranostics 2022, 6, 286–305. [Google Scholar] [CrossRef]
  330. Trebeschi, S.; Drago, S.; Birkbak, N.; Kurilova, I.; Cǎlin, A.; Pizzi, A.D.; Lalezari, F.; Lambregts, D.; Rohaan, M.; Parmar, C.; et al. Predicting response to cancer immunotherapy using noninvasive radiomic biomarkers. Ann. Oncol. 2019, 30, 998–1004. [Google Scholar] [CrossRef] [Green Version]
  331. O’Connor, J.P.B.; Aboagye, E.; Adams, J.E.; Aerts, H.J.W.L.; Barrington, S.F.; Beer, A.J.; Boellaard, R.; Bohndiek, S.; Brady, M.; Brown, G.; et al. Imaging biomarker roadmap for cancer studies. Nat. Rev. Clin. Oncol. 2017, 14, 169–186. [Google Scholar] [CrossRef]
  332. Im, Y.; Tsui, D.; Diaz, L.; Wan, J. Next-Generation Liquid Biopsies: Embracing Data Science in Oncology. Trends Cancer 2021, 7, 283–292. [Google Scholar] [CrossRef] [PubMed]
  333. Yang, X.-L.; Shi, Y.; Zhang, D.-D.; Xin, R.; Deng, J.; Wu, T.-M.; Wang, H.-M.; Wang, P.-Y.; Liu, J.-B.; Li, W.; et al. Quantitative proteomics characterization of cancer biomarkers and treatment. Mol. Ther.-Oncolytics 2021, 21, 255–263. [Google Scholar] [CrossRef] [PubMed]
  334. Davis-Marcisak, E.F.; Deshpande, A.; Stein-O’Brien, G.L.; Ho, W.J.; Laheru, D.; Jaffee, E.M.; Fertig, E.J.; Kagohara, L.T. From bench to bedside: Single-cell analysis for cancer immunotherapy. Cancer Cell 2021, 39, 1062–1080. [Google Scholar] [CrossRef] [PubMed]
  335. Newell, F.; da Silva, I.P.; Johansson, P.A.; Menzies, A.M.; Wilmott, J.S.; Addala, V.; Carlino, M.S.; Rizos, H.; Nones, K.; Edwards, J.J.; et al. Multiomic profiling of checkpoint inhibitor-treated melanoma: Identifying predictors of response and resistance, and markers of biological discordance. Cancer Cell 2022, 40, 88–102.e7. [Google Scholar] [CrossRef]
  336. Menetski, J.P.; Austin, C.P.; Brady, L.S.; Eakin, G.; Leptak, C.; Meltzer, A.; Wagner, J.A. The FNIH Biomarkers Consortium embraces the BEST. Nat. Rev. Drug Discov. 2019, 18, 567–568. [Google Scholar] [CrossRef] [PubMed]
  337. Mullard, A. $215 million cancer immunotherapy biomarker consortium debuts. Nat. Rev. Drug Discov. 2017, 16, 743. [Google Scholar] [CrossRef]
  338. Oh, D.Y.; Fong, L.; Newell, E.W.; Turk, M.J.; Chi, H.; Chang, H.Y.; Satpathy, A.T.; Fairfax, B.; Silva-Santos, B.; Lantz, O. Toward a better understanding of T cells in cancer. Cancer Cell 2021, 39, 1549–1552. [Google Scholar] [CrossRef]
  339. Sharma, P.; Siddiqui, B.A.; Anandhan, S.; Yadav, S.S.; Subudhi, S.K.; Gao, J.; Goswami, S.; Allison, J.P. The Next Decade of Immune Checkpoint Therapy. Cancer Discov. 2021, 11, 838–857. [Google Scholar] [CrossRef]
  340. Yost, K.E.; Satpathy, A.T.; Wells, D.K.; Qi, Y.; Wang, C.; Kageyama, R.; McNamara, K.L.; Granja, J.M.; Sarin, K.Y.; Brown, R.A.; et al. Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat. Med. 2019, 25, 1251–1259. [Google Scholar] [CrossRef]
  341. Chowell, D.; Yoo, S.-K.; Valero, C.; Pastore, A.; Krishna, C.; Lee, M.; Hoen, D.; Shi, H.; Kelly, D.W.; Patel, N.; et al. Improved prediction of immune checkpoint blockade efficacy across multiple cancer types. Nat. Biotechnol. 2021, 40, 499–506. [Google Scholar] [CrossRef]
  342. Ignatiadis, M.; Sledge, G.W.; Jeffrey, S.S. Liquid biopsy enters the clinic—Implementation issues and future challenges. Nat. Rev. Clin. Oncol. 2021, 18, 297–312. [Google Scholar] [CrossRef] [PubMed]
Table 1. Clinical and technical challenges in the use of PD-L1+CTCs as a biomarker for anti-PD-(L)1 immunotherapy.
Table 1. Clinical and technical challenges in the use of PD-L1+CTCs as a biomarker for anti-PD-(L)1 immunotherapy.
RationaleClinical ChallengesTechnical ChallengesRecent Advances and Trends
  • Role of CTCs in tumor progression and resistance to anti-PD-(L)1 immunotherapy [71,72]
  • CTCs from tumor tissues can be detected in most cancer patients and have been shown to be associated with prognosis [73]
  • The number of PD-L1+CTCs is associated with the clinical efficacy of anti-PD-(L)1 immunotherapy [75,76]
  • The patient enrollment size for clinical studies is small
  • Not validated by randomized clinical trials using a validated PD-L1+CTCs testing platform
  • Clinical trial data are from different lines and different anti-PD-(L)1 antibodies [15,97,98]
  • Heterogeneity of CTC populations and very low amount of CTCs in circulating blood [93,94,95]
  • Lack of robustness and accuracy in isolating sufficient numbers of CTCs [90,95,97,98]
  • Testing PD-L1+CTCs from different cell/tissue sources [97,101,102]
  • Testing for post-translational modifications of PD-L1 [103,104,105,106]
  • Testing for the PD-L1 positive CTCs/immune cell clusters [111,112]
  • Combination with other biomarkers
Table 2. Clinical and technical challenges in the use of PD-L1-positive circulating immune cells as a biomarker for anti-PD-(L)1 immunotherapy.
Table 2. Clinical and technical challenges in the use of PD-L1-positive circulating immune cells as a biomarker for anti-PD-(L)1 immunotherapy.
RationaleClinical ChallengesTechnical ChallengesRecent Advances and Trends
  • All types of immune cells participate in tumor development, progression, metastasis, and resistance to anti-cancer therapies including anti-PD-(L)1 immunotherapy [39,127,128,129,130,131,132,133]
  • Systemic immunity is critical for immune-mediated tumor eradication [9,39]
  • PBMCs contain both tumor-infiltrating immune cells as well as immune cells activated in the periphery by anti-PD-(L)1 immunotherapy [142,143,144]
  • PD-L1 positive immune cells from tumor tissues can be detected in blood [136,137,138]
  • The patient enrollment size of clinical studies is small
  • Not validated by randomized clinical trials using validated PD-L1+CTCs testing platform
  • The biology of PD-L1 on various subsets of immune cells remains not well understood
  • Conflicting predictions of PD-L1 expression as a biomarker on a variety of immune cells’ response to anti-PD-(L)1 immunotherapy
  • Lack of standard platform to capture immune cell complexity
  • Heterogeneity of immune cells, as well as dynamic temporal changes of PD-L1 ex-pression on immune cells [266,267,268,269,270,271]
  • Novel flow cytometry-based assay [263,264]
  • Artificial intelligence for diagnostic and machine learning techniques [266,267,268,269,270,271]
  • Combining PD-L (1) positive immune cells with other biomarkers
Table 3. Clinical and technical challenges in the use of cePD-L1 as a biomarker for anti-PD-(L)1 immunotherapy.
Table 3. Clinical and technical challenges in the use of cePD-L1 as a biomarker for anti-PD-(L)1 immunotherapy.
RationaleClinical ChallengesTechnical ChallengesRecent Advances and Trends
  • Derive from all cell types in the primary tumor or metastatic tissues [274]
  • Participate in every stage of cancer development and in modulating anti-cancer therapies [275,276,277,278,279,280,281,282,283]
  • PD-L1 is highly expressed on tumor-derived exosomes and functions similarly to PD-L1 expressed on tumor cells and immune cells [284,285,286]
  • The levels of exoPD-L1 are associated with clinical outcome [287,288]
  • Gene expression patterns between tumor tissues and exosomes detected in plasma or serum are closely correlated [68,289]
  • The patient enrollment size of clinical studies is small
  • Not validated by randomized clinical trials using validated PD-L1+CTCs testing platform
  • No correlation between exoPD-L1 and PD-L1 assessed by IHC in tumors [288,297]
  • Inconsistent data on the correlation of exo/cePD-L1 and response to anti-PD-(L)1 immunotherapies [285,288,292]
  • Conflicting studies on the expression of exo/cePD-(L)1 [66,283,290]
  • Lack of standardized isolation and purification methods for circulating exosomes [291,292]
  • Characterization of exosomes from different compenents of tumor tissues [292]
  • Combination of exo/cePD-(L)1 with other biomarkers [280,288,289]
  • Technologies to quantifiy exo/cePD-L1 [278,279,286,295]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wang, T.; Denman, D.; Bacot, S.M.; Feldman, G.M. Challenges and the Evolving Landscape of Assessing Blood-Based PD-L1 Expression as a Biomarker for Anti-PD-(L)1 Immunotherapy. Biomedicines 2022, 10, 1181. https://doi.org/10.3390/biomedicines10051181

AMA Style

Wang T, Denman D, Bacot SM, Feldman GM. Challenges and the Evolving Landscape of Assessing Blood-Based PD-L1 Expression as a Biomarker for Anti-PD-(L)1 Immunotherapy. Biomedicines. 2022; 10(5):1181. https://doi.org/10.3390/biomedicines10051181

Chicago/Turabian Style

Wang, Tao, Desirée Denman, Silvia M. Bacot, and Gerald M. Feldman. 2022. "Challenges and the Evolving Landscape of Assessing Blood-Based PD-L1 Expression as a Biomarker for Anti-PD-(L)1 Immunotherapy" Biomedicines 10, no. 5: 1181. https://doi.org/10.3390/biomedicines10051181

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