Biomarkers in Non-Small Cell Lung Cancer: Comparison
Please note this is a comparison between Version 2 by Jason Zhu and Version 1 by Katiane Tostes.

Lung cancer has the highest mortality rate among all cancer types, resulting in over 1.8 million deaths annually. Immunotherapy utilizing immune checkpoint inhibitors (ICIs) has revolutionized the treatment of non-small cell lung cancer (NSCLC). ICIs, predominantly monoclonal antibodies, modulate co-stimulatory and co-inhibitory signals crucial for maintaining immune tolerance. Despite significant therapeutic advancements in NSCLC, patients still face challenges such as disease progression, recurrence, and high mortality rates. Therefore, there is a need for predictive biomarkers that can guide lung cancer treatment strategies. 

  • immunotherapy
  • immune checkpoint inhibitors
  • non-small cell lung cancer

1. Introduction

While immunomodulatory agents have shown promising results in treating refractory solid tumors [42] [1, not all patients exhibit satisfactory responses. Immunotoxicity is a common occurrence, underscoring the need for predictive biomarkers to identify patients who would benefit from immunotherapy [43] [2. Furthermore, the high cost of immunotherapies poses a barrier to access, limiting this treatment option to a privileged subset of patients in low–middle-income countries.

2. PD-L1 Expression

PD-L1 expression is the currently used biomarker for predicting the response and clinical efficacy of ICIs in NSCLC [44] [3. Positive PD-L1 expression is associated with better outcomes and clinical response rates than those for patients with negative PD-L1 negative expression [45] [4, regardless of smoking habits [46] [5. PD-L1 expression levels can be measured using the Tumor Proportion Score (TPS) or Combined Positive Score (CPS), where TPS represents the percentage of tumor cells expressing PD-L1 and CPS accounts for PD-L1 presence in both tumor and inflammatory cells. Pro-inflammatory signals associated with JAK2 transduction [47] [6 and IFN-γ expression [48] [7 may increase PD-L1 expression [49] [8.
In a phase I study, PD-L1 surface expression levels > 5% were associated with a response to nivolumab, while patients with PD-L1 expression < 5% did not respond well to treatment [50] [9. In the phase III Keynote 024 study, patients with aNSCLC and high levels of PD-L1 expression (≥50%) demonstrated improved outcomes when treated with pembrolizumab compared to the platinum-based CT group [39] [10. Although patients with advanced squamous cell NSCLC showed better responses to nivolumab than to docetaxel regardless of PD-L1 expression, even patients with negative PD-L1 expression experienced increased PFS after ICI treatment [35] [11.
However, despite these findings, PD-L1 TPS alone is not sufficiently accurate or reliable in predicting the response to immunotherapy [51] [12. The technical limitations of immunohistochemistry (IHC) in the context of PD-L1 expression analysis include variations in sample quality, as the measurements are conducted on tumor biopsies that may not fully capture tumor heterogeneity. Additionally, diverse staining protocols and the establishment of appropriate cut-off points for interpreting PD-L1 expression pose further challenges [52] [13. In addition, the predictive capacity of IHC in determining the response to ICIs across various histologies, such as SCC, remains limited [34,35] [11][14. In this context, the combination of TPS and CPS is a more robust biomarker [53] [15.
PD-L1 identification in circulating tumor cells (CTCs) tends to be higher than in tissues, potentially enhancing quantification efficiency. However, results regarding its correlation with treatment response remain controversial, with studies reporting absence of or worse-lasting response to ICIs with higher pretreatment PD-L1+ CTCs, while others indicate the positive prognostic value of PD-L1 expression in CTCs [54,55,56] [16][17][18. Some studies found no correlation between PD-L1 expression in tumor biopsies and CTCs [57] [19.
Tumor cells that do not express PD-L1 may still be accompanied by immune cells expressing this ligand. Additionally, tumor-infiltrating immune cells in the tumor environment demonstrate increased levels of PD-L1 expression, and the administration of ICI leads to an enhanced influx of CD8+ T lymphocytes in the tumor region [58] [20.
Plasma soluble PD-L1 (sPD-L1) levels have been investigated as an alternative to PD-L1 TPS. While certain studies have found no correlation between pre-treatment levels of soluble programmed death-ligand 1 (sPD-L1) and OS [59] [21, other investigations have established a significant association between elevated pre-treatment sPD-L1 levels and unfavorable outcomes, including higher rates of treatment failure [60] [22. Furthermore, increased or stable levels of soluble PD-1 (sPD-1) after two cycles of nivolumab were linked to a better prognosis [61] [23.
Genetic variations were also evaluated as possible biomarkers. PD-L1 polymorphisms were associated with the efficacy of ICIs, being correlated with OS improvement [62] [24. Patients carrying HER2 exon 20 [63] [25, ERBB4 [64] [26, KRAS G12C [65] [27, FGFR4 [66] [28, ARID 1A, and ARID 1B [67] [29 mutations showed ICIs benefit. However, patients harboring KRAS G12V [65] [27, EGFR [68] [30, or ALK [69] [31 alterations tend to have poorer outcomes.

3. Gene-Expression-Based Biomarkers

Gene-expression-based biomarkers have been successfully employed in the field of oncology for the clinical management of cancer patients [72] [32. In patients treated with anti-PD-1, the expression levels of CSF1 R and HCST positively correlated with PD-L1 levels and high infiltration of CD8+ T cells [73] [33. Nevertheless, other potential ICI response signatures have been described, including the expression levels of MAP1A/1B/1S/4/6/7D1/7D3 in ADC and SCC, where high expression of these genes was associated with favorable response to immunotherapy [74] [34. In NSCLC, KAT2B expression displayed a positive correlation with the levels of infiltrating immune cells and mRNA expression of immune checkpoint genes. Conversely, tumor tissues exhibited downregulation of KAT2B expression, which was associated with ineffective response to ICI and unfavorable prognosis in patients with lung ADC [75] [35.
A TCR co-expression signature has been identified as a valuable predictor of prognosis in NSCLC patients undergoing immunotherapy. Elevated expression levels of this specific gene-set are indicative of more favorable treatment responses [76] [36. Additionally, a cancer-specific immune score model has been developed to predict ICI response with satisfactory performance, achieving an area under the curve (AUC) of 0.68, indicating its potential for accurately assessing ICI response [77] [37.
Moreover, the gene expression profile within the TME holds predictive value for pathologic complete response and disease progression in patients receiving combined neoadjuvant chemoimmunotherapy. Tumors showing favorable response to treatment exhibited elevated levels of IFNG, GZMB, NKG7, and M1 macrophages whereas tumors prone to relapse following surgery exhibited heightened expression of genes such as AKT1, BST2, OAS3, and CD8B [78] [38.
To construct an immune gene score, pivotal immune cells, human leukocyte antigens (HLAs), and immune checkpoints were selected and the immune-related genes of those three aspects of the TME were combined to construct the score. The score derived from these three aspects demonstrated the ability to predict the response to ICIs, achieving an area under the curve (AUC) of 0.737 at the 20-month mark. Remarkably, within this signature, patients exhibiting a higher hypoxia score demonstrated a stronger association with immunotherapy efficacy. A predictive model integrating this immune score, tumor mutational burden (TMB), and long non-coding RNA expression exhibited promising predictive potential for effective immunotherapy response, achieving an AUC of 0.814 at 20 months [79] [39.
Efforts are ongoing to identify gene-expression-based signatures that can accurately predict response to immunotherapy. However, current studies are in the preliminary stages and require further validation before their potential for clinical implementation can be fully assessed [80] [40.

4. Tumor Mutation Burden

TMB refers to the presence of non-synonymous mutations in the coding regions of the tumor genome [81] [41. High TMB indicates a greater number of neoantigens, which can activate the T-lymphocyte response [82] [42. In NSCLC, the mutagenic effects of smoking can lead to higher TMB rates [83] [43. Several studies have correlated TMB rates with response to ICIs [82 [42][44,84], suggesting that a TMB of ten or more mutations per megabase is associated with improved PFS [85] [45. Patients treated with pembrolizumab and high TMB rates have demonstrated durable clinical benefits [86] [46. Additionally, in ADC patients, a high burden of clonal neoantigens has been linked to better outcomes and ICI responses [87] [47.
However, it is worth noting that less than 10% of nonsynonymous mutations result in immunogenicity [88] [48. Additionally, there is currently no established cut-off point for non-synonymous mutations that can reliably predict clinical benefit [89] [49. In colorectal cancer (CRC) patients, elevated TMB levels and microsatellite instability (MSI) are associated with a more favorable prognosis [90] [50. However, the prevalence of MSI-high (MSI-H) status in NSCLC patients is rare, limiting its usefulness as a biomarker in this patient population [91] [51. Despite these limitations, the combination of TMB with other biomarkers such as PD-L1 expression or the neutrophil-to-lymphocyte ratio (NLR) holds promise, as it enhances prediction capabilities [92] [52.

5. Complete Blood Count

Absolute complete blood count (CBC) values have been evaluated as potential biomarkers for predicting the response to ICIs by analyzing patients’ medical records [93] [53. In NSCLC, the presence of local inflammation results in an immune infiltrate rich in neutrophils [94] [54. The increase in NLR [95 [55][56][57,96,97], myeloid-to-lymphoid ratio (M:L), absolute-neutrophil-count-to-absolute-lymphocyte-count ratio (ANC:ALC) and absolute neutrophil counts (post-ANCs) is associated with lower PFS [98] [58 and OS [99] [59. Conversely, low platelet-to-lymphocyte ratio (PLR) [96 [56][60,100], monocyte-to-lymphocyte ratio (MLR) [96] [56, and high rates of ALC [101] [61, absolute eosinophil count (AEC) [102] [62, and relative eosinophil count (REC) [103] [63 are associated with better PFS.
Low levels of hemoglobin (HGB) [104] [64, red blood cell (RBC) counts, and hematocrit (HCT), which reflect anemia, are associated with shorter OS. In this case, the ICI response can be identified by the combination of NLR and HGB [95] [55. The Lung Immune Prognostic Index (LIPI) score, which takes into account the derived neutrophil-to-lymphocyte ratio (dNLR) and lactate dehydrogenase level (LDH), is also associated with OS, with patients scoring zero demonstrating favorable outcomes [85] [45. However, studies have demonstrated that high levels of dNLR are present in patients who experience early failure with ICI [105] [65.
Peripheral blood biomarkers, particularly those obtained from routine examinations, represent a significant advancement in clinical practice [106] [66. Nonetheless, further investigation is necessary to fully understand and validate these findings.

6. Peripheral Blood Mononuclear Cells

Peripheral blood mononuclear cells (PBMCs) encompass various immune cell types, including monocytes, T cells, B cells, granulocytes, and natural killer cells (NK), and they play a crucial role in the initial immune defense against malignancies [108] [67. Regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) are implicated in tumor growth and exert immunosuppressive functions in cancer patients [109] [68.
In NSCLC patients treated with anti-PD-1 therapy, responders exhibit increased proliferation of T PD-1+CD8+cells with an effector phenotype (HLA-DR+ CD38+ BCL-2low) and elevated expression of CD28 [110] [69. High proliferation of PD-1+CD8+ T cells [111] [70, PD-L1 expressing CD14+ monocytes [112] [71 and Forkhead Box P3 (FoxP3+) Treg cells [113] [72 may also be associated with treatment response. However, an augmented frequency of TIM-3+ T lymphocytes, whether CD4+ or CD8+ T cells, negatively correlates to PFS [114] [73. Likewise, high levels of CCR9+ or CCR10+CD4+ T cells or CXCR4+CD8+ T cells negatively correlate with ICI treatment survival outcomes [115] [74.
Nivolumab-treated NSCLC patients with a high central memory/effector T cell ratio demonstrate prolonged PFS and higher tumor PD-L1 expression [116] [75. Furthermore, an increase in exhausted cells (TIGIT+) and a decrease in memory effector CD8+ T cells (CCR7 CD45RA) are associated with disease progression [117] [76.
The ratio of Tregs to Lox-1+ PMN-MDSCs (TMR) can be employed after the initial immunotherapy infusion and exhibits higher sensitivity in predicting negative treatment responses [109] [68. In contrast, high levels of granulocytic myeloid-derived suppressor cells (Gr-MDSC) are related to a positive response to treatment with ICIs [97] [57. Nevertheless, further studies are necessary to elucidate these conflicting findings.

7. Tumor-Infiltrating Immune Cells

The TME comprises non-malignant stromal cells, bone-marrow-derived cells, and tumor-infiltrating lymphocytes (TILs) [119] [77. In resume, the TME immune population of consists of macrophages, dendritic cells, natural killer cells, B cells, effector T helper cells, and Treg and cytotoxic T cells [120] [78.
Evidence suggests that PD-L1-positive NSCLC patients treated with ICIs who have stromal CD8+ effector T cells as the most abundant TIL subpopulation experience better outcomes (Table 6). However, TILs are distributed heterogeneously, and their predictive value may be diminished in patients expressing multiple markers of T cell exhaustion [121] [79, limiting their association with treatment outcomes.
The measurement of TIL populations using computational methods also shows promise, as the spatial distribution of TILs may be linked to ICI response [122,123] [80][81. Studies have classified cohorts into three main phenotypes based on TME inflammation: immune-inflamed, immune-excluded, and immune-deserted. Among these, immune-inflamed phenotypes exhibit better responses to ICIs. Compared to other biomarkers, TIL levels can be more predictive than TMB load for PD-L1-negative patients [124] [82.
Proteins have also been investigated as potential biomarkers for treatment response. Examples include CD24, which co-stimulates clonal expansion of CD4 T-cells [125,126] [83][84; CD73, which is involved in lymphocyte differentiation [127,128] [85][86; and CD137, which is associated with cancer immunity [129,130] [87][88. However, CD24 positivity has been correlated with worse PFS in PD-L1 < 50% patients treated with ICIs based on IHC analyses [125] [83. In addition, the increase in CD73 and CD137 correlates with better PFS [129] [87, regardless of PD-L1 status [127] [85, but these results are still controversial [131] [89. The major limitation of this methodology is the scarcity of NSCLC tumor tissue [52] [13.

8. Extracellular Vesicles

Extracellular vesicles (EVs), which includes exosomes and microvesicles, play a crucial role in the cellular communication mechanism by transporting bioactive molecules that can influence the extracellular environment and the immune system [132] [90. EVs derived from tumor tissue have the potential to serve as non-invasive biomarkers due to their molecular composition, which reflects the complexity and heterogeneity of the tumor microenvironment [133] [91. Some molecules carried by EVs, such as PD-L1, TGF-β1, FasL, TRAIL, COX2, CD39/CD73, CTLA4, and NKG2D, are involved in tumor evasion and immunosuppression, and therefore hold promise as predictive biomarkers for immunotherapy response [134] [92.
Studies have shown that responders to ICIs exhibit higher levels of tetraspanin co-stimulatory molecules, specifically CD9, CD81, and CD63, in EVs. These findings suggest that these molecules may serve as promising biomarkers associated with a better objective response rate (ORR) [135] [93. Conversely, another study identified the overexpression of TGF-β in EVs as a predictor of poorer outcomes and non-response to ICI treatment [136] [94.
EVs can carry small molecules, such as microRNAs (miRNAs), that are widely studied and can also present predictive value [137] [95. For instance, EV-miR-625-5p has been described as an independent biomarker of response to ICIs in NSCLC patients with PD-L1 expression ≥50% [138] [96. Furthermore, pre-treatment concentrations of EVs with an endothelial phenotype (CD41a/CD31+/CD45) in the blood have been correlated with longer OS, PFS, and clinical response to ICIs. Proteomic analysis has revealed that responders to ICIs have distinct protein loading in EVs at baseline and during treatment [139] [97.
In terms of conventional biomarkers for immune response, EVs offer an alternative method for measuring PD-L1 levels [140,141] [98][99. Studies show that increased exosomal PD-L1 expression is associated with better ORR, OS, and treatment efficacy [140] [98. However, elevated levels of extracellular vesicles are more common in non-responders [139 [97][99,141], although they may not always predict a sustained response or survival [141] [99. A novel biochip has been proposed to quantify PD-1/PD-L1 proteins on the surface of extracellular vesicles and EV PD-1/PD-L1 mRNA (Au SERP). This tool has shown 72.2% accuracy in detecting ICI responders and non-responders [142] [100.

9. Imaging Biomarkers

Medical imaging techniques, such as positron emission tomography-computed tomography (PET-CT), can provide insights into the cellular and molecular properties of tumors. These scans have been used to correlate the expression of PD-1/PD-L1 and the response to treatment [143,144,145,146] [101][102][103][104. Quantifications occur by detecting 89Zr probes that are linked to monoclonal antibodies (mAbs) administered during immunotherapy. Studies utilizing 89Zr-nivolumab [145] [103 and 89Zr-atezolizumab [143] [101 particles have demonstrated their effectiveness in predicting the response to ICIs. On the other hand, 89Zr-durvalumab [146] [104, although considered safe and feasible, only exhibited a weak correlation with treatment response.
The application of positron emission tomography-computed tomography using 18F-fluorodeoxyglucose (18F-FDG PET-CT) has shown promise in determining the metabolic tumor volume (tMTV), which serves as a reliable predictor of pembrolizumab efficacy. Furthermore, tMTV may be a valuable tool for guiding treatment decisions in patients who require more aggressive therapeutic interventions, such as chemoimmunotherapy [144] [102.

10. Microbiome

The respiratory tract microbiome has demonstrated its potential to predict the response to ICI in NSCLC patients [147,148,149] [105][106][107. A dysbiotic signature in the respiratory tract microbiome has been associated with tumor progression and a poorer prognosis [150] [108. Conversely, a more diverse lung microbiome is linked to higher levels of CXCL9, a chemokine associated with better ICI response [149] [107.
Moreover, the application of 16S RNA sequencing has identified specific microbial enrichments in NSCLC patients with different ICI responses. Poor responders to ICIs showed an enrichment of Fusobacterium nucleatum [147] [105, Haemophilus influenzae, and Neisseria perflava [148] [106. In contrast, patients with an enrichment of Veillonella dispar [148] [106 and Akkermansia muciniphila (AKK) [151] [109 demonstrated a favorable response to ICIs.
The use of antibiotics can alter the gut microbiome and is correlated with unfavorable clinical outcomes, as patients treated with antibiotics exhibit lower alpha diversity [152] [110. Evidence suggests that antibiotic treatment reduces the presence of CXCL9 in the lung tumor microenvironment, which may be associated with lower sensitivity to ICIs [149] [107. Further studies are needed to fully understand the intricate interactions within the cancer–microbiome–immune axis.

References

  1. Moskovitz, J.M.; Ferris, R.L. Tumor Immunology and Immunotherapy for Head and Neck Squamous Cell Carcinoma. J. Dent. Res. 2018, 97, 622–626.
  2. Bodor, J.N.; Boumber, Y.; Borghaei, H. Biomarkers for immune checkpoint inhibition in non-small cell lung cancer (NSCLC). Cancer 2020, 126, 260–270.
  3. Jeanson, A.; Tomasini, P.; Souquet-Bressand, M.; Brandone, N.; Boucekine, M.; Grangeon, M.; Chaleat, S.; Khobta, N.; Milia, J.; Mhanna, L.; et al. Efficacy of Immune Checkpoint Inhibitors in KRAS-Mutant Non-Small Cell Lung Cancer (NSCLC). J. Thorac. Oncol. 2019, 14, 1095–1101.
  4. Doroshow, D.B.; Wei, W.; Gupta, S.; Zugazagoitia, J.; Robbins, C.; Adamson, B.; Rimm, D.L. Programmed Death-Ligand 1 Tumor Proportion Score and Overall Survival From First-Line Pembrolizumab in Patients With Nonsquamous Versus Squamous NSCLC. J. Thorac. Oncol. 2021, 16, 2139–2143.
  5. Gainor, J.F.; Rizvi, H.; Jimenez Aguilar, E.; Skoulidis, F.; Yeap, B.Y.; Naidoo, J.; Khosrowjerdi, S.; Mooradian, M.; Lydon, C.; Illei, P.; et al. Clinical activity of programmed cell death 1 (PD-1) blockade in never, light, and heavy smokers with non-small-cell lung cancer and PD-L1 expression >/=50. Ann. Oncol. 2020, 31, 404–411.
  6. Green, M.R.; Monti, S.; Rodig, S.J.; Juszczynski, P.; Currie, T.; O’Donnell, E.; Chapuy, B.; Takeyama, K.; Neuberg, D.; Golub, T.R.; et al. Integrative analysis reveals selective 9p24.1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma. Blood 2010, 116, 3268–3277.
  7. Liu, J.; Hamrouni, A.; Wolowiec, D.; Coiteux, V.; Kuliczkowski, K.; Hetuin, D.; Saudemont, A.; Quesnel, B. Plasma cells from multiple myeloma patients express B7-H1 (PD-L1) and increase expression after stimulation with IFN-gamma and TLR ligands via a MyD88-, TRAF6-, and MEK-dependent pathway. Blood 2007, 110, 296–304.
  8. Mazzaschi, G.; Madeddu, D.; Falco, A.; Bocchialini, G.; Goldoni, M.; Sogni, F.; Armani, G.; Lagrasta, C.A.; Lorusso, B.; Mangiaracina, C.; et al. Low PD-1 Expression in Cytotoxic CD8(+) Tumor-Infiltrating Lymphocytes Confers an Immune-Privileged Tissue Microenvironment in NSCLC with a Prognostic and Predictive Value. Clin. Cancer Res. 2018, 24, 407–419.
  9. Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B.; et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 2012, 366, 2443–2454.
  10. Reck, M.; Rodriguez-Abreu, D.; Robinson, A.G.; Hui, R.; Csoszi, T.; Fulop, A.; Gottfried, M.; Peled, N.; Tafreshi, A.; Cuffe, S.; et al. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2016, 375, 1823–1833.
  11. Brahmer, J.; Reckamp, K.L.; Baas, P.; Crino, L.; Eberhardt, W.E.; Poddubskaya, E.; Antonia, S.; Pluzanski, A.; Vokes, E.E.; Holgado, E.; et al. Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2015, 373, 123–135.
  12. Zou, Y.; Hu, X.; Zheng, S.; Yang, A.; Li, X.; Tang, H.; Kong, Y.; Xie, X. Discordance of immunotherapy response predictive biomarkers between primary lesions and paired metastases in tumours: A systematic review and meta-analysis. EBioMedicine 2021, 63, 103137.
  13. Liberini, V.; Mariniello, A.; Righi, L.; Capozza, M.; Delcuratolo, M.D.; Terreno, E.; Farsad, M.; Volante, M.; Novello, S.; Deandreis, D. NSCLC Biomarkers to Predict Response to Immunotherapy with Checkpoint Inhibitors (ICI): From the Cells to In Vivo Images. Cancers 2021, 13, 4543.
  14. Borghaei, H.; Paz-Ares, L.; Horn, L.; Spigel, D.R.; Steins, M.; Ready, N.E.; Chow, L.Q.; Vokes, E.E.; Felip, E.; Holgado, E.; et al. Nivolumab versus Docetaxel in Advanced Nonsquamous Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2015, 373, 1627–1639.
  15. Herbst, R.S.; Soria, J.C.; Kowanetz, M.; Fine, G.D.; Hamid, O.; Gordon, M.S.; Sosman, J.A.; McDermott, D.F.; Powderly, J.D.; Gettinger, S.N.; et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014, 515, 563–567.
  16. 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.
  17. Tamminga, M.; de Wit, S.; Hiltermann, T.J.N.; Timens, W.; Schuuring, E.; Terstappen, L.; Groen, H.J.M. 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.
  18. Dall’Olio, F.G.; Gelsomino, F.; Conci, N.; Marcolin, L.; De Giglio, A.; Grilli, G.; Sperandi, F.; Fontana, F.; Terracciano, M.; Fragomeno, B.; et al. PD-L1 Expression in Circulating Tumor Cells as a Promising Prognostic Biomarker in Advanced Non-small-cell Lung Cancer Treated with Immune Checkpoint Inhibitors. Clin. Lung Cancer 2021, 22, 423–431.
  19. Janning, M.; Kobus, F.; Babayan, A.; Wikman, H.; Velthaus, J.L.; Bergmann, S.; Schatz, S.; Falk, M.; Berger, L.A.; Bottcher, L.M.; et al. Determination of PD-L1 Expression in Circulating Tumor Cells of NSCLC Patients and Correlation with Response to PD-1/PD-L1 Inhibitors. Cancers 2019, 11, 835.
  20. Forde, P.M.; Chaft, J.E.; Smith, K.N.; Anagnostou, V.; Cottrell, T.R.; Hellmann, M.D.; Zahurak, M.; Yang, S.C.; Jones, D.R.; Broderick, S.; et al. Neoadjuvant PD-1 Blockade in Resectable Lung Cancer. N. Engl. J. Med. 2018, 378, 1976–1986.
  21. Ando, K.; Hamada, K.; Watanabe, M.; Ohkuma, R.; Shida, M.; Onoue, R.; Kubota, Y.; Matsui, H.; Ishiguro, T.; Hirasawa, Y.; et al. Plasma Levels of Soluble PD-L1 Correlate With Tumor Regression in Patients With Lung and Gastric Cancer Treated With Immune Checkpoint Inhibitors. Anticancer Res. 2019, 39, 5195–5201.
  22. Mazzaschi, G.; Minari, R.; Zecca, A.; Cavazzoni, A.; Ferri, V.; Mori, C.; Squadrilli, A.; Bordi, P.; Buti, S.; Bersanelli, M.; et al. Soluble PD-L1 and Circulating CD8+PD-1+ and NK Cells Enclose a Prognostic and Predictive Immune Effector Score in Immunotherapy Treated NSCLC patients. Lung Cancer 2020, 148, 1–11.
  23. Tiako Meyo, M.; Jouinot, A.; Giroux-Leprieur, E.; Fabre, E.; Wislez, M.; Alifano, M.; Leroy, K.; Boudou-Rouquette, P.; Tlemsani, C.; Khoudour, N.; et al. Predictive Value of Soluble PD-1, PD-L1, VEGFA, CD40 Ligand and CD44 for Nivolumab Therapy in Advanced Non-Small Cell Lung Cancer: A Case-Control Study. Cancers 2020, 12, 473.
  24. Yoshida, H.; Nomizo, T.; Ozasa, H.; Tsuji, T.; Funazo, T.; Yasuda, Y.; Ajimizu, H.; Yamazoe, M.; Kuninaga, K.; Ogimoto, T.; et al. PD-L1 polymorphisms predict survival outcomes in advanced non-small-cell lung cancer patients treated with PD-1 blockade. Eur. J. Cancer 2021, 144, 317–325.
  25. Lau, S.C.M.; Fares, A.F.; Le, L.W.; Mackay, K.M.; Soberano, S.; Chan, S.W.; Smith, E.; Ryan, M.; Tsao, M.S.; Bradbury, P.A.; et al. Subtypes of EGFR- and HER2-Mutant Metastatic NSCLC Influence Response to Immune Checkpoint Inhibitors. Clin. Lung Cancer 2021, 22, 253–259.
  26. Hu, X.; Xu, H.; Xue, Q.; Wen, R.; Jiao, W.; Tian, K. The role of ERBB4 mutations in the prognosis of advanced non-small cell lung cancer treated with immune checkpoint inhibitors. Mol. Med. 2021, 27, 126.
  27. Wu, S.G.; Liao, W.Y.; Su, K.Y.; Yu, S.L.; Huang, Y.L.; Yu, C.J.; Chih-Hsin Yang, J.; Shih, J.Y. Prognostic Characteristics and Immunotherapy Response of Patients With Nonsquamous NSCLC With Kras Mutation in East Asian Populations: A Single-Center Cohort Study in Taiwan. JTO Clin. Res. Rep. 2021, 2, 100140.
  28. Wang, L.; Ren, Z.; Yu, B.; Tang, J. Development of nomogram based on immune-related gene FGFR4 for advanced non-small cell lung cancer patients with sensitivity to immune checkpoint inhibitors. J. Transl. Med. 2021, 19, 22.
  29. Sun, D.; Tian, L.; Zhu, Y.; Wo, Y.; Liu, Q.; Liu, S.; Li, H.; Hou, H. Subunits of ARID1 serve as novel biomarkers for the sensitivity to immune checkpoint inhibitors and prognosis of advanced non-small cell lung cancer. Mol. Med. 2020, 26, 78.
  30. Lee, C.K.; Man, J.; Lord, S.; Links, M.; Gebski, V.; Mok, T.; Yang, J.C. Checkpoint Inhibitors in Metastatic EGFR-Mutated Non-Small Cell Lung Cancer-A Meta-Analysis. J. Thorac. Oncol. 2017, 12, 403–407.
  31. Nishio, M.; Takahashi, T.; Yoshioka, H.; Nakagawa, K.; Fukuhara, T.; Yamada, K.; Ichiki, M.; Tanaka, H.; Seto, T.; Sakai, H.; et al. KEYNOTE-025: Phase 1b study of pembrolizumab in Japanese patients with previously treated programmed death ligand 1-positive advanced non-small-cell lung cancer. Cancer Sci. 2019, 110, 1012–1020.
  32. Soliman, H.; Shah, V.; Srkalovic, G.; Mahtani, R.; Levine, E.; Mavromatis, B.; Srinivasiah, J.; Kassar, M.; Gabordi, R.; Qamar, R.; et al. MammaPrint guides treatment decisions in breast Cancer: Results of the IMPACt trial. BMC Cancer 2020, 20, 81.
  33. Qi, X.; Qi, C.; Wu, T.; Hu, Y. CSF1R and HCST: Novel Candidate Biomarkers Predicting the Response to Immunotherapy in Non-Small Cell Lung Cancer. Technol. Cancer Res. Treat. 2020, 19, 1533033820970663.
  34. Luo, J.; Hu, Q.; Gou, M.; Liu, X.; Qin, Y.; Zhu, J.; Cai, C.; Tian, T.; Tu, Z.; Du, Y.; et al. Expression of Microtubule-Associated Proteins in Relation to Prognosis and Efficacy of Immunotherapy in Non-Small Cell Lung Cancer. Front. Oncol. 2021, 11, 680402.
  35. Zhou, X.; Wang, N.; Zhang, Y.; Yu, H.; Wu, Q. KAT2B is an immune infiltration-associated biomarker predicting prognosis and response to immunotherapy in non-small cell lung cancer. Investig. New Drugs 2022, 40, 43–57.
  36. Wang, Y.; Liu, Y.; Li, X.; Li, W.; Xue, Z.; He, X.; Xiong, W.; He, L.; Bai, Y. TCR Coexpression Signature Predicts Immunotherapy Resistance in NSCLC. Front. Pharmacol. 2022, 13, 875149.
  37. Das, S.; Camphausen, K.; Shankavaram, U. Cancer-Specific Immune Prognostic Signature in Solid Tumors and Its Relation to Immune Checkpoint Therapies. Cancers 2020, 12, 2476.
  38. Casarrubios, M.; Provencio, M.; Nadal, E.; Insa, A.; Del Rosario Garcia-Campelo, M.; Lazaro-Quintela, M.; Domine, M.; Majem, M.; Rodriguez-Abreu, D.; Martinez-Marti, A.; et al. Tumor microenvironment gene expression profiles associated to complete pathological response and disease progression in resectable NSCLC patients treated with neoadjuvant chemoimmunotherapy. J. Immunother. Cancer 2022, 10, e005320.
  39. Lin, L.; Zhang, W.; Chen, Y.; Ren, W.; Zhao, J.; Ouyang, W.; He, Z.; Su, W.; Yao, H.; Yu, Y. Immune gene patterns and characterization of the tumor immune microenvironment associated with cancer immunotherapy efficacy. Heliyon 2023, 9, e14450.
  40. Ma, Y.; Yang, J.; Ji, T.; Wen, F. Identification of a novel m5C/m6A-related gene signature for predicting prognosis and immunotherapy efficacy in lung adenocarcinoma. Front. Genet. 2022, 13, 990623.
  41. Snyder, A.; Makarov, V.; Merghoub, T.; Yuan, J.; Zaretsky, J.M.; Desrichard, A.; Walsh, L.A.; Postow, M.A.; Wong, P.; Ho, T.S.; et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 2014, 371, 2189–2199.
  42. Kim, E.S.; Velcheti, V.; Mekhail, T.; Yun, C.; Shagan, S.M.; Hu, S.; Chae, Y.K.; Leal, T.A.; Dowell, J.E.; Tsai, M.L.; et al. Blood-based tumor mutational burden as a biomarker for atezolizumab in non-small cell lung cancer: The phase 2 B-F1RST trial. Nat. Med. 2022, 28, 939–945.
  43. Alexandrov, L.B.; Nik-Zainal, S.; Wedge, D.C.; Aparicio, S.A.; Behjati, S.; Biankin, A.V.; Bignell, G.R.; Bolli, N.; Borg, A.; Borresen-Dale, A.L.; et al. Signatures of mutational processes in human cancer. Nature 2013, 500, 415–421.
  44. Peters, S.; Dziadziuszko, R.; Morabito, A.; Felip, E.; Gadgeel, S.M.; Cheema, P.; Cobo, M.; Andric, Z.; Barrios, C.H.; Yamaguchi, M.; et al. Atezolizumab versus chemotherapy in advanced or metastatic NSCLC with high blood-based tumor mutational burden: Primary analysis of BFAST cohort C randomized phase 3 trial. Nat. Med. 2022, 28, 1831–1839.
  45. Ali, W.A.S.; Hui, P.; Ma, Y.; Wu, Y.; Zhang, Y.; Chen, Y.; Hong, S.; Yang, Y.; Huang, Y.; Zhao, Y.; et al. Determinants of survival in advanced non-small cell lung cancer patients treated with anti-PD-1/PD-L1 therapy. Ann. Transl. Med. 2021, 9, 1639.
  46. Rizvi, N.A.; Hellmann, M.D.; Snyder, A.; Kvistborg, P.; Makarov, V.; Havel, J.J.; Lee, W.; Yuan, J.; Wong, P.; Ho, T.S.; et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015, 348, 124–128.
  47. McGranahan, N.; Furness, A.J.; Rosenthal, R.; Ramskov, S.; Lyngaa, R.; Saini, S.K.; Jamal-Hanjani, M.; Wilson, G.A.; Birkbak, N.J.; Hiley, C.T.; et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016, 351, 1463–1469.
  48. Yadav, M.; Jhunjhunwala, S.; Phung, Q.T.; Lupardus, P.; Tanguay, J.; Bumbaca, S.; Franci, C.; Cheung, T.K.; Fritsche, J.; Weinschenk, T.; et al. Predicting immunogenic tumour mutations by combining mass spectrometry and exome sequencing. Nature 2014, 515, 572–576.
  49. Pradhan, M.; Chocry, M.; Gibbons, D.L.; Sepesi, B.; Cascone, T. Emerging biomarkers for neoadjuvant immune checkpoint inhibitors in operable non-small cell lung cancer. Transl. Lung Cancer Res. 2021, 10, 590–606.
  50. Berardinelli, G.N.; Duraes, R.; Mafra da Costa, A.; Bragagnoli, A.; Antonio de Oliveira, M.; Pereira, R.; Scapulatempo-Neto, C.; Guimaraes, D.P.; Reis, R.M. Association of microsatellite instability (MSI) status with the 5-year outcome and genetic ancestry in a large Brazilian cohort of colorectal cancer. Eur. J. Hum. Genet. 2022, 30, 824–832.
  51. De Marchi, P.; Berardinelli, G.N.; Cavagna, R.O.; Pinto, I.A.; da Silva, F.A.F.; Duval da Silva, V.; Santana, I.V.V.; da Silva, E.C.A.; Ferro Leal, L.; Reis, R.M. Microsatellite Instability Is Rare in the Admixed Brazilian Population of Non-Small Cell Lung Cancer: A Cohort of 526 Cases. Pathobiology 2022, 89, 101–106.
  52. Kao, C.; Powers, E.; Wu, Y.; Datto, M.B.; Green, M.F.; Strickler, J.H.; Ready, N.E.; Zhang, T.; Clarke, J.M. Predictive Value of Combining Biomarkers for Clinical Outcomes in Advanced Non-Small Cell Lung Cancer Patients Receiving Immune Checkpoint Inhibitors. Clin. Lung Cancer 2021, 22, 500–509.
  53. Laza-Briviesca, R.; Cruz-Bermudez, A.; Nadal, E.; Insa, A.; Garcia-Campelo, M.D.R.; Huidobro, G.; Domine, M.; Majem, M.; Rodriguez-Abreu, D.; Martinez-Marti, A.; et al. Blood biomarkers associated to complete pathological response on NSCLC patients treated with neoadjuvant chemoimmunotherapy included in NADIM clinical trial. Clin. Transl. Med. 2021, 11, e491.
  54. Singel, K.L.; Segal, B.H. Neutrophils in the tumor microenvironment: Trying to heal the wound that cannot heal. Immunol. Rev. 2016, 273, 329–343.
  55. 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.
  56. Sanchez-Gastaldo, A.; Munoz-Fuentes, M.A.; Molina-Pinelo, S.; Alonso-Garcia, M.; Boyero, L.; Bernabe-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.
  57. 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.
  58. Kubo, S.; Kobayashi, N.; Somekawa, K.; Hirata, M.; Kamimaki, C.; Aiko, H.; Katakura, S.; Teranishi, S.; Watanabe, K.; Hara, Y.U.; et al. Identification of Biomarkers for Non-small-cell Lung Cancer Patients Treated with an Immune Checkpoint Inhibitor. Anticancer Res. 2020, 40, 3889–3896.
  59. 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.
  60. Jiang, M.; Peng, W.; Pu, X.; Chen, B.; Li, J.; Xu, F.; Liu, L.; Xu, L.; Xu, Y.; Cao, J.; et al. Peripheral Blood Biomarkers Associated with Outcome in Non-small Cell Lung Cancer Patients Treated with Nivolumab and Durvalumab Monotherapy. Front. Oncol. 2020, 10, 913.
  61. Huemer, F.; Lang, D.; Westphal, T.; Gampenrieder, S.P.; Hutarew, G.; Weiss, L.; Hackl, H.; Lamprecht, B.; Rinnerthaler, G.; Greil, R. Baseline Absolute Lymphocyte Count and ECOG Performance Score Are Associated with Survival in Advanced Non-Small Cell Lung Cancer Undergoing PD-1/PD-L1 Blockade. J. Clin. Med. 2019, 8, 1014.
  62. Caliman, E.; Fancelli, S.; Ottanelli, C.; Mazzoni, F.; Paglialunga, L.; Lavacchi, D.; Michelet, M.R.G.; Giommoni, E.; Napolitano, B.; Scolari, F.; et al. Absolute eosinophil count predicts clinical outcomes and toxicity in non-small cell lung cancer patients treated with immunotherapy. Cancer Treat. Res. Commun. 2022, 32, 100603.
  63. Sibille, A.; Henket, M.; Corhay, J.L.; Alfieri, R.; Louis, R.; Duysinx, B. White Blood Cells in Patients Treated with Programmed Cell Death-1 Inhibitors for Non-small Cell Lung Cancer. Lung 2021, 199, 549–557.
  64. Zhang, Z.; Zhang, F.; Yuan, F.; Li, Y.; Ma, J.; Ou, Q.; Liu, Z.; Yang, B.; Wang, L.; Tao, H.; et al. Pretreatment hemoglobin level as a predictor to evaluate the efficacy of immune checkpoint inhibitors in patients with advanced non-small cell lung cancer. Ther. Adv. Med. Oncol. 2020, 12, 1758835920970049.
  65. Alessi, J.V.; Ricciuti, B.; Alden, S.L.; Bertram, A.A.; Lin, J.J.; Sakhi, M.; Nishino, M.; Vaz, V.R.; Lindsay, J.; Turner, M.M.; et al. Low peripheral blood derived neutrophil-to-lymphocyte ratio (dNLR) is associated with increased tumor T-cell infiltration and favorable outcomes to first-line pembrolizumab in non-small cell lung cancer. J. Immunother. Cancer 2021, 9, e003536.
  66. You, W.; Shang, B.; Sun, J.; Liu, X.; Su, L.; Jiang, S. Mechanistic insight of predictive biomarkers for antitumor PD-1/PD-L1 blockade: A paradigm shift towards immunome evaluation (Review). Oncol. Rep. 2020, 44, 424–437.
  67. Rotunno, M.; Hu, N.; Su, H.; Wang, C.; Goldstein, A.M.; Bergen, A.W.; Consonni, D.; Pesatori, A.C.; Bertazzi, P.A.; Wacholder, S.; et al. A gene expression signature from peripheral whole blood for stage I lung adenocarcinoma. Cancer Prev. Res. 2011, 4, 1599–1608.
  68. Kim, H.R.; Park, S.M.; Seo, S.U.; Jung, I.; Yoon, H.I.; Gabrilovich, D.I.; Cho, B.C.; Seong, S.Y.; Ha, S.J.; Youn, J.I. The Ratio of Peripheral Regulatory T Cells to Lox-1(+) Polymorphonuclear Myeloid-derived Suppressor Cells Predicts the Early Response to Anti-PD-1 Therapy in Patients with Non-Small Cell Lung Cancer. Am. J. Respir. Crit. Care Med. 2019, 199, 243–246.
  69. Kamphorst, A.O.; Pillai, R.N.; Yang, S.; Nasti, T.H.; Akondy, R.S.; Wieland, A.; Sica, G.L.; Yu, K.; Koenig, L.; Patel, N.T.; et al. Proliferation of PD-1+ CD8 T cells in peripheral blood after PD-1-targeted therapy in lung cancer patients. Proc. Natl. Acad. Sci. USA 2017, 114, 4993–4998.
  70. Ferrara, R.; Naigeon, M.; Auclin, E.; Duchemann, B.; Cassard, L.; Jouniaux, J.M.; Boselli, L.; Grivel, J.; Desnoyer, A.; Mezquita, L.; et al. Circulating T-cell Immunosenescence in Patients with Advanced Non-small Cell Lung Cancer Treated with Single-agent PD-1/PD-L1 Inhibitors or Platinum-based Chemotherapy. Clin. Cancer Res. 2021, 27, 492–503.
  71. 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.
  72. 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.
  73. Julia, E.P.; Mando, P.; Rizzo, M.M.; Cueto, G.R.; Tsou, F.; Luca, R.; Pupareli, C.; Bravo, A.I.; Astorino, W.; Mordoh, J.; et al. Peripheral changes in immune cell populations and soluble mediators after anti-PD-1 therapy in non-small cell lung cancer and renal cell carcinoma patients. Cancer Immunol. Immunother. 2019, 68, 1585–1596.
  74. Rogado, J.; Pozo, F.; Troule, K.; Sanchez-Torres, J.M.; Romero-Laorden, N.; Mondejar, R.; Donnay, O.; Ballesteros, A.; Pacheco-Barcia, V.; Aspa, J.; et al. Peripheral Blood Mononuclear Cells Predict Therapeutic Efficacy of Immunotherapy in NSCLC. Cancers 2022, 14, 2898.
  75. Manjarrez-Orduno, N.; Menard, L.C.; Kansal, S.; Fischer, P.; Kakrecha, B.; Jiang, C.; Cunningham, M.; Greenawalt, D.; Patel, V.; Yang, M.; et al. Circulating T Cell Subpopulations Correlate with Immune Responses at the Tumor Site and Clinical Response to PD1 Inhibition in Non-Small Cell Lung Cancer. Front. Immunol. 2018, 9, 1613.
  76. Kim, C.G.; Kim, K.H.; Pyo, K.H.; Xin, C.F.; Hong, M.H.; Ahn, B.C.; Kim, Y.; Choi, S.J.; Yoon, H.I.; Lee, J.G.; et al. Hyperprogressive disease during PD-1/PD-L1 blockade in patients with non-small-cell lung cancer. Ann. Oncol. 2019, 30, 1104–1113.
  77. Chen, F.; Zhuang, X.; Lin, L.; Yu, P.; Wang, Y.; Shi, Y.; Hu, G.; Sun, Y. New horizons in tumor microenvironment biology: Challenges and opportunities. BMC Med. 2015, 13, 45.
  78. Yang, L.; Pang, Y.; Moses, H.L. TGF-beta and immune cells: An important regulatory axis in the tumor microenvironment and progression. Trends Immunol. 2010, 31, 220–227.
  79. Lopez de Rodas, M.; Nagineni, V.; Ravi, A.; Datar, I.J.; Mino-Kenudson, M.; Corredor, G.; Barrera, C.; Behlman, L.; Rimm, D.L.; Herbst, R.S.; et al. Role of tumor infiltrating lymphocytes and spatial immune heterogeneity in sensitivity to PD-1 axis blockers in non-small cell lung cancer. J. Immunother. Cancer 2022, 10, e004440.
  80. Park, S.; Ock, C.Y.; Kim, H.; Pereira, S.; Park, S.; Ma, M.; Choi, S.; Kim, S.; Shin, S.; Aum, B.J.; et al. Artificial Intelligence-Powered Spatial Analysis of Tumor-Infiltrating Lymphocytes as Complementary Biomarker for Immune Checkpoint Inhibition in Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2022, 40, 1916–1928.
  81. Wang, X.; Barrera, C.; Bera, K.; Viswanathan, V.S.; Azarianpour-Esfahani, S.; Koyuncu, C.; Velu, P.; Feldman, M.D.; Yang, M.; Fu, P.; et al. Spatial interplay patterns of cancer nuclei and tumor-infiltrating lymphocytes (TILs) predict clinical benefit for immune checkpoint inhibitors. Sci. Adv. 2022, 8, eabn3966.
  82. Rakaee, M.; Adib, E.; Ricciuti, B.; Sholl, L.M.; Shi, W.; Alessi, J.V.; Cortellini, A.; Fulgenzi, C.A.M.; Viola, P.; Pinato, D.J.; et al. Association of Machine Learning-Based Assessment of Tumor-Infiltrating Lymphocytes on Standard Histologic Images with Outcomes of Immunotherapy in Patients with NSCLC. JAMA Oncol. 2023, 9, 51–60.
  83. Ozawa, Y.; Harutani, Y.; Oyanagi, J.; Akamatsu, H.; Murakami, E.; Shibaki, R.; Hayata, A.; Sugimoto, T.; Tanaka, M.; Takakura, T.; et al. CD24, not CD47, negatively impacts upon response to PD-1/L1 inhibitors in non-small-cell lung cancer with PD-L1 tumor proportion score < 50. Cancer Sci. 2021, 112, 72–80.
  84. Fang, X.; Zheng, P.; Tang, J.; Liu, Y. CD24: From A to Z. Cell. Mol. Immunol. 2010, 7, 100–103.
  85. Ishii, H.; Azuma, K.; Kawahara, A.; Kinoshita, T.; Matsuo, N.; Naito, Y.; Tokito, T.; Yamada, K.; Akiba, J.; Hoshino, T. Predictive value of CD73 expression for the efficacy of immune checkpoint inhibitors in NSCLC. Thorac. Cancer 2020, 11, 950–955.
  86. Zhang, B. CD73: A novel target for cancer immunotherapy. Cancer Res. 2010, 70, 6407–6411.
  87. Hwang, S.; Kwon, A.Y.; Jeong, J.Y.; Kim, S.; Kang, H.; Park, J.; Kim, J.H.; Han, O.J.; Lim, S.M.; An, H.J. Immune gene signatures for predicting durable clinical benefit of anti-PD-1 immunotherapy in patients with non-small cell lung cancer. Sci. Rep. 2020, 10, 643.
  88. Glorieux, C.; Huang, P. CD137 expression in cancer cells: Regulation and significance. Cancer Commun. 2019, 39, 70.
  89. Ramdani, H.O.; Falk, M.; Heukamp, L.C.; Schatz, S.; Tiemann, M.; Wesseler, C.; Diehl, L.; Schuuring, E.; Groen, H.J.M.; Griesinger, F. Immune related endonucleases and GTPases are not associated with tumor response in patients with advanced non-small cell lung cancer treated with checkpoint inhibitors. Pathol. Res. Pract. 2021, 227, 153651.
  90. Becker, A.; Thakur, B.K.; Weiss, J.M.; Kim, H.S.; Peinado, H.; Lyden, D. Extracellular Vesicles in Cancer: Cell-to-Cell Mediators of Metastasis. Cancer Cell 2016, 30, 836–848.
  91. Ruivo, C.F.; Adem, B.; Silva, M.; Melo, S.A. The Biology of Cancer Exosomes: Insights and New Perspectives. Cancer Res. 2017, 77, 6480–6488.
  92. Wolf, G.T.; Moyer, J.S.; Kaplan, M.J.; Newman, J.G.; Egan, J.E.; Berinstein, N.L.; Whiteside, T.L. IRX-2 natural cytokine biologic for immunotherapy in patients with head and neck cancers. OncoTargets Ther. 2018, 11, 3731–3746.
  93. Signorelli, D.; Ghidotti, P.; Proto, C.; Brambilla, M.; De Toma, A.; Ferrara, R.; Galli, G.; Ganzinelli, M.; Lo Russo, G.; Prelaj, A.; et al. Circulating CD81-expressing extracellular vesicles as biomarkers of response for immune-checkpoint inhibitors in advanced NSCLC. Front. Immunol. 2022, 13, 987639.
  94. de Miguel-Perez, D.; Russo, A.; Gunasekaran, M.; Buemi, F.; Hester, L.; Fan, X.; Carter-Cooper, B.A.; Lapidus, R.G.; Peleg, A.; Arroyo-Hernandez, M.; et al. Baseline extracellular vesicle TGF-beta is a predictive biomarker for response to immune checkpoint inhibitors and survival in non-small cell lung cancer. Cancer 2023, 129, 521–530.
  95. Durendez-Saez, E.; Torres-Martinez, S.; Calabuig-Farinas, S.; Meri-Abad, M.; Ferrero-Gimeno, M.; Camps, C. Exosomal microRNAs in non-small cell lung cancer. Transl. Cancer Res. 2021, 10, 3128–3139.
  96. Pantano, F.; Zalfa, F.; Iuliani, M.; Simonetti, S.; Manca, P.; Napolitano, A.; Tiberi, S.; Russano, M.; Citarella, F.; Foderaro, S.; et al. Large-Scale Profiling of Extracellular Vesicles Identified miR-625-5p as a Novel Biomarker of Immunotherapy Response in Advanced Non-Small-Cell Lung Cancer Patients. Cancers 2022, 14, 2435.
  97. Brocco, D.; Lanuti, P.; Pieragostino, D.; Cufaro, M.C.; Simeone, P.; Bologna, G.; Di Marino, P.; De Tursi, M.; Grassadonia, A.; Irtelli, L.; et al. Phenotypic and Proteomic Analysis Identifies Hallmarks of Blood Circulating Extracellular Vesicles in NSCLC Responders to Immune Checkpoint Inhibitors. Cancers 2021, 13, 585.
  98. 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.
  99. de Miguel-Perez, D.; Russo, A.; Arrieta, O.; Ak, M.; Barron, F.; Gunasekaran, M.; Mamindla, P.; Lara-Mejia, L.; Peterson, C.B.; Er, M.E.; et al. Extracellular vesicle PD-L1 dynamics predict durable response to immune-checkpoint inhibitors and survival in patients with non-small cell lung cancer. J. Exp. Clin. Cancer Res. 2022, 41, 186.
  100. Nguyen, L.T.H.; Zhang, J.; Rima, X.Y.; Wang, X.; Kwak, K.J.; Okimoto, T.; Amann, J.; Yoon, M.J.; Shukuya, T.; Chiang, C.L.; et al. An immunogold single extracellular vesicular RNA and protein ((Au) SERP) biochip to predict responses to immunotherapy in non-small cell lung cancer patients. J. Extracell. Vesicles 2022, 11, e12258.
  101. Bensch, F.; van der Veen, E.L.; Lub-de Hooge, M.N.; Jorritsma-Smit, A.; Boellaard, R.; Kok, I.C.; Oosting, S.F.; Schroder, C.P.; Hiltermann, T.J.N.; van der Wekken, A.J.; et al. (89)Zr-atezolizumab imaging as a non-invasive approach to assess clinical response to PD-L1 blockade in cancer. Nat. Med. 2018, 24, 1852–1858.
  102. Dall’Olio, F.G.; Calabro, D.; Conci, N.; Argalia, G.; Marchese, P.V.; Fabbri, F.; Fragomeno, B.; Ricci, D.; Fanti, S.; Ambrosini, V.; et al. Baseline total metabolic tumour volume on 2-deoxy-2-fluoro-d-glucose positron emission tomography-computed tomography as a promising biomarker in patients with advanced non-small cell lung cancer treated with first-line pembrolizumab. Eur. J. Cancer 2021, 150, 99–107.
  103. Niemeijer, A.N.; Leung, D.; Huisman, M.C.; Bahce, I.; Hoekstra, O.S.; van Dongen, G.; Boellaard, R.; Du, S.; Hayes, W.; Smith, R.; et al. Whole body PD-1 and PD-L1 positron emission tomography in patients with non-small-cell lung cancer. Nat. Commun. 2018, 9, 4664.
  104. Smit, J.; Borm, F.J.; Niemeijer, A.N.; Huisman, M.C.; Hoekstra, O.S.; Boellaard, R.; Oprea-Lager, D.E.; Vugts, D.J.; van Dongen, G.; de Wit-van der Veen, B.J.; et al. PD-L1 PET/CT Imaging with Radiolabeled Durvalumab in Patients with Advanced-Stage Non-Small Cell Lung Cancer. J. Nucl. Med. 2022, 63, 686–693.
  105. Chu, S.; Cheng, Z.; Yin, Z.; Xu, J.; Wu, F.; Jin, Y.; Yang, G. Airway Fusobacterium is Associated with Poor Response to Immunotherapy in Lung Cancer. OncoTargets Ther. 2022, 15, 201–213.
  106. Jang, H.J.; Choi, J.Y.; Kim, K.; Yong, S.H.; Kim, Y.W.; Kim, S.Y.; Kim, E.Y.; Jung, J.Y.; Kang, Y.A.; Park, M.S.; et al. Relationship of the lung microbiome with PD-L1 expression and immunotherapy response in lung cancer. Respir. Res. 2021, 22, 322.
  107. Masuhiro, K.; Tamiya, M.; Fujimoto, K.; Koyama, S.; Naito, Y.; Osa, A.; Hirai, T.; Suzuki, H.; Okamoto, N.; Shiroyama, T.; et al. Bronchoalveolar lavage fluid reveals factors contributing to the efficacy of PD-1 blockade in lung cancer. JCI Insight 2022, 7, e157915.
  108. Tsay, J.J.; Wu, B.G.; Sulaiman, I.; Gershner, K.; Schluger, R.; Li, Y.; Yie, T.A.; Meyn, P.; Olsen, E.; Perez, L.; et al. Lower Airway Dysbiosis Affects Lung Cancer Progression. Cancer Discov. 2021, 11, 293–307.
  109. Derosa, L.; Routy, B.; Thomas, A.M.; Iebba, V.; Zalcman, G.; Friard, S.; Mazieres, J.; Audigier-Valette, C.; Moro-Sibilot, D.; Goldwasser, F.; et al. Intestinal Akkermansia muciniphila predicts clinical response to PD-1 blockade in patients with advanced non-small-cell lung cancer. Nat. Med. 2022, 28, 315–324.
  110. Hakozaki, T.; Richard, C.; Elkrief, A.; Hosomi, Y.; Benlaifaoui, M.; Mimpen, I.; Terrisse, S.; Derosa, L.; Zitvogel, L.; Routy, B.; et al. The Gut Microbiome Associates with Immune Checkpoint Inhibition Outcomes in Patients with Advanced Non-Small Cell Lung Cancer. Cancer Immunol. Res. 2020, 8, 1243–1250.
More
Video Production Service