Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2632 2022-03-31 11:10:45 |
2 format correct Meta information modification 2632 2022-04-01 05:36:43 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Ushio, R.; Murakami, S.; , . Predictive Markers for Immune Checkpoint Inhibitors. Encyclopedia. Available online: https://encyclopedia.pub/entry/21208 (accessed on 24 April 2024).
Ushio R, Murakami S,  . Predictive Markers for Immune Checkpoint Inhibitors. Encyclopedia. Available at: https://encyclopedia.pub/entry/21208. Accessed April 24, 2024.
Ushio, Ryota, Shuji Murakami,  . "Predictive Markers for Immune Checkpoint Inhibitors" Encyclopedia, https://encyclopedia.pub/entry/21208 (accessed April 24, 2024).
Ushio, R., Murakami, S., & , . (2022, March 31). Predictive Markers for Immune Checkpoint Inhibitors. In Encyclopedia. https://encyclopedia.pub/entry/21208
Ushio, Ryota, et al. "Predictive Markers for Immune Checkpoint Inhibitors." Encyclopedia. Web. 31 March, 2022.
Predictive Markers for Immune Checkpoint Inhibitors
Edit

Immune checkpoint inhibitors (ICIs) have dramatically improved the outcomes of non-small cell lung cancer patients and have increased the possibility of long-term survival. However, few patients benefit from ICIs, and no predictive biomarkers other than tumor programmed cell death ligand 1 (PD-L1) expression have been established. Hence, the identification of biomarkers is an urgent issue.

non-small cell lung cancer biomarker anti-programmed cell death ligand 1

1. Introduction

Lung cancer is the most frequent cause of cancer death worldwide. In 2020, 2.21 million new cases (11.4% of all cancer cases) and 1.80 million deaths (18.0% of all cancer deaths) were reported [1]. The most common histological type is non-small cell lung cancer (NSCLC), and most patients are diagnosed at an advanced stage [2]. Platinum-based chemotherapy has historically been the standard treatment for NSCLC, although limited therapeutic effects in patients with a poor prognosis have been observed. Recently, the advent of immune checkpoint inhibitors (ICIs) such as nivolumab and pembrolizumab (anti-programmed cell death 1 (PD-1) antibodies), atezolizumab and durvalumab (anti-programmed cell death ligand 1 (PD-L1) antibodies), and ipilimumab and tremelimumab (anti-cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) antibody) has dramatically altered the approach of advanced NSCLC treatment. First-line ICI therapy has demonstrated more prolonged survival than conventional platinum-based chemotherapy for stage IV NSCLC. In a phase III trial (KEYNOTE-024), pembrolizumab increased overall survival (OS) to 30 months for NSCLC patients with a PD-L1 tumor proportion score (TPS) > 50%, thereby demonstrating its superiority to conventional platinum-based chemotherapy [3]. Furthermore, a phase III trial (KEYNOTE-042) comparing pembrolizumab monotherapy with platinum-based combination therapy in NSCLC patients with PD-L1 TPS ≥ 1% showed significantly longer OS in the pembrolizumab group than in the chemotherapy group [4]. Atezolizumab also prolonged OS over platinum-based chemotherapy (17.5 vs. 15.1 months) in advanced NSCLC with PD-L1 ≥ 1% of tumor cells (TCs) or ≥ 1% of tumor-infiltrating immune cells (ICs), regardless of histology [5]. In a phase III study (CheckMate 227), nivolumab plus ipilimumab was associated with better OS than chemotherapy (17.1 vs. 14.9 months) in patients with NSCLC, regardless of PD-L1 expression level [6]. First-line ICIs combined with chemotherapy are among the current standard therapies for advanced NSCLC, compensating for the disadvantages of early treatment failure with ICI monotherapy. In a phase III trial (KEYNOTE-189) of patients with advanced NSCLC, the addition of pembrolizumab to platinum-doublet chemotherapy significantly prolonged PFS and OS (survival rate at 12 months was 69.2% vs. 49.4%) [7]. In addition, this combination therapy overcame the early failure of ICIs, which had been a problem with single-agent therapy [7]. In a phase III trial (KEYNOTE-407) evaluating the efficacy of pembrolizumab added to platinum-based combination therapy in patients with advanced lung squamous cell carcinoma (LUSC), OS was significantly prolonged (17.1 vs. 11.6 months) [8]. A phase III study (IMpower150) revealed that the addition of atezolizumab to carboplatin/paclitaxel or carboplatin/paclitaxel/bevacizumab in non-squamous NSCLC patients significantly prolonged OS to 19.2 months [9]. Furthermore, in another phase III study (IMpower130), atezolizumab added to carboplatin/nab-paclitaxel significantly prolonged OS to 18.6 months [10], while in the CheckMate 9LA phase III trial, nivolumab/ipilimumab in combination with platinum-based therapy significantly prolonged OS to 15.6 months compared with platinum-based therapy in NSCLC patients [11]. ICIs also raised the possibility that advanced NSCLC patients may have a better chance of long-term survival. A first-line phase III immunotherapy trial (KEYNOTE-024) with pembrolizumab for NSCLC achieved a 5-year OS rate of 31.9% [12]. Two phase III trials of nivolumab (CheckMate 017 and CheckMate 057) in patients with previously treated advanced NSCLC demonstrated 5-year OS rates of 13.4% and 8.0%, respectively [13].
However, many NSCLC patients do not benefit from ICIs or suffer significant life-threatening immunotoxicity [14]. Immune-related adverse events (ir-AEs) can affect various organs, and dermatitis, pneumonitis, colitis, and endocrinopathies tend to be most common. While most cases are mild to moderate in severity, some cases are severe or even fatal, especially when not promptly recognized and appropriately managed [15][16]. Although it is essential to administer ICIs to appropriate patients, the expression of programmed death-ligand 1 (PD-L1), a widely used biomarker, is not a sufficient predictive factor. ICIs are effective even in NSCLC patients with low or absent PD-L1 expression and may not be effective in patients with high PD-L1 expression. Therefore, there is an urgent need to identify new biomarkers to predict the response to ICIs for selecting the best anti-cancer agents for each patient.

2. Predictive Biomarkers beyond PD-L1 Expression

2.1. Tumor Mutational Burden

Tumor mutational burden (TMB) is defined by the number of mutation calls (somatic single variant (SNV) and multinucleotide variant (MNV) and small insertions and deletions (indels)) per megabase (Mb) of interrogated coding sequences. These mutations can be transcribed and translated into neoantigen-containing peptides, processed by the antigen-processing machinery, and loaded onto major histocompatibility complex (MHC) molecules for presentation on the cell surface. The immune system recognizes neoantigens as non-self-immunogenic targets, activating and targeting T cells [17][18][19][20]. Tissue and blood TMB is a potential biomarker of immunotherapy outcomes in multiple tumor types. In particular, lung cancer is primarily caused by chronic exposure to carcinogens in cigarette smoke, and the efficacy of ICIs correlates with a molecular signature characteristic of cigarette carcinogen-related mutagenesis, certain DNA repair mutations, and the burden of neoantigens [21][22]. An analysis of the CheckMate 568 study of nivolumab plus ipilimumab in NSCLC reported that ORR increased in patients with a higher tissue tumor mutational burden (tTMB) using the FoundationOne CDx (F1CDx) assay, plateaued at a threshold of 10 mutations (mut)/Mb (ORR: 4%, 10%, 44%, and 39% in patients with TMB <5, <10, 10, and ≥15 mut/Mb, respectively), and the enhanced response was independent of PD-L1 expression [23]. The CheckMate 227 study of nivolumab plus ipilimumab in NSCLC also demonstrated longer PFS in patients with tTMB-high, with at least 10 mut/Mb, irrespective of tumor PD-L1 expression level [24], while a phase III trial of durvalumab (MYSTIC) and tremelimumab indicated longer PFS and OS in NSCLC patients with blood TMB (bTMB)-high, with at least 20 mut/Mb [21]. A retrospective analysis of the POPLAR and OAK studies demonstrated that a positive correlation of tTMB and bTMB and NSCLC patients with bTMB ≥ 16 mut/Mb led to an increased PFS benefit from atezolizumab [25].
Although TMB is optimally calculated by whole-exome sequencing (WES), this approach presents difficulties in terms of its substantial cost and turnaround time in clinical settings [26]. To address this, targeted sequencing assays enriched with known cancer-driving gene mutations are used to assess TMB. The F1CDx assay and Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT) assay are moderately concordant with WES in TMB analysis [27][28], and both assays were recently approved as companion diagnostics by the FDA to assess TMB in solid tumors. In this approval, tTMB-high was defined as having at least 10 mut/Mb according to the KEYNOTE-158 study of pembrolizumab for unresectable or metastatic solid tumors [29]. Despite these initial positive findings, the role of TMB as a biomarker in NSCLC remains unclear. It was reported that tTMB was not predictive of the efficacy of pembrolizumab alone or in combination with chemotherapy according to retrospective analyses of the KEYNOTE-189 and KEYNOTE-021 studies, respectively [30][31].
TMB as a biomarker has other limitations, including a lack of standardization between the testing platforms. Low tumor purity may lead to inaccurate TMB estimates [32]. Lung cancer specimens often have a low tumor cell content due to inflammatory cells and stromal components, leading to an underestimation of TMB. Furthermore, although high-TMB is thought to lead to increased neoantigens, the effect on the tumor immune response may vary depending on whether the neoantigen is derived from clonal (or homogeneous tumor) or subclonal (or heterogeneous tumor) mutations because the lower antigen dosage compared with the clonal neoantigen burden reduces the chances of identifying T cells reactive to subclonal neoantigens. McGranahan et al. demonstrated that sensitivity to ICIs in NSCLC and melanoma patients was enhanced in tumors enriched for clonal neoantigens, and cytotoxic chemotherapy-induced subclonal neoantigens were enriched in certain poor responders [33].

2.2. DNA Mismatch Repair Deficiency and Microsatellite Instability

DNA mismatch repair (MMR) is a highly conserved biological DNA repair pathway in mammalian cells and is crucial for maintaining genomic stability. MMR deficiency (dMMR) is the initiating event in many cancer types [34]. The deficient DNA MMR mechanism leads to missed DNA replication errors, resulting in the increased acquisition of mutations, primarily in the form of microsatellite instability (MSI) or alterations in microsatellites, which increases the burden of neoantigens [35][36].
A clinical trial of pembrolizumab across dMMR tumors spanning 12 cancer types demonstrated that ORR was 53%, and complete response (CR) was 21% [37]. This led to pan-cancer approval by the FDA. However, the role of the MMR status as a predictive biomarker for immunotherapy in lung cancer remains unknown because it was not included in this study. The prevalence of MSI-high (MSI-H) status is rare, at 0.53% and 0.60% of lung adenocarcinomas (LUAD) and LUSCs, respectively [38].

2.3. CD8+ Tumor-Infiltrating Lymphocytes

The adaptive immune system identifies and targets tumor cells. Interestingly, CD8+ T cells, CD4+ T cells, B cells, dendritic cells, and effectors of innate immunity, namely macrophages, polymorphonuclear leukocytes, and natural killer cells (NK), as well as all cell types within the tumor, are classified as tumor-infiltrating lymphocytes (TILs) [39][40]. Of them all, the presence of tumor-infiltrating CD8+ T cells, which recognize tumor antigens, is a prerequisite for successful ICI treatment when presented at the tumor cell surface in the context of HLA class I. Several small-sized studies and a meta-analysis demonstrated that CD8+ TILs were significantly associated with better OS, PFS, and ORR in NSCLC patients treated with ICIs [41][42][43][44][45][46][47]. Shirasawa et al. demonstrated a classification system based on PD-L1 expression and CD8+ TIL status that accurately predicts the efficacy of ICIs in NSCLC patients better than tumor PD-L1 expression [48]. Furthermore, Kumagami et al. showed that the frequency of PD-1+CD8+ T cells relative to that of PD-1+ regulatory T (Treg) cells in the tumor microenvironment could predict the efficacy of ICIs more accurately than tumor PD-L1 expression [49].
Although TILs have great predictive power, they present some technical problems as biomarkers in clinical practice, and CD8+ TILs within the stroma and invasive margin compartment indicate a better outcome than those in the intratumoral compartment [45][50]. However, biopsy samples obtained by CT-guided needle biopsy or bronchoscopy in patients with advanced NSCLC are often insufficient to evaluate stromal TILs, and a sample may not represent the TME of the entire primary tumor or metastatic lesions.

2.4. Human Leukocyte Antigen Class I

The human leukocyte antigen (HLA) system encodes cell-surface proteins involved in immune system regulation [51]. Furthermore, HLA-I presents peptides derived from intracellular proteins on the surface of CD8+ T cells, so that cancer cells are killed [52][53].
Several specific HLA-I genotypes are suggested as biomarkers for ICI treatment; Naranbhai et al. reported that HLA-A*03 alleles led to a low ORR and poor PFS and OS in various cancer patients, including NSCLC with ICI treatment, in the most significant epidemiological analysis of the association between HLA-I and ICI efficacy so far [54]. In melanoma patients, HLA-B44 supertype and HLA-A02 supertype led to prolonged OS with ICI treatment [55][56]. Losses in heterozygosity (LOH) and HLA gene expression have also been reported as candidate biomarkers. Chowell et al. reported that LOH in cancer reduces OS in NSCLC and melanoma patients with ICI treatment [55][56]. However, Negrao et al. reported no significant correlations between HLA-I zygosity and PFS or OS in NSCLC patients with ICI treatment [57]. Schaafsma et al. analyzed 33 cancer types and reported that tumors with high HLA gene expression tended to have higher immune cell infiltration, including CD8+ T and NK cells, and a more immunologically active TME, thus leading to increased survival [58].

2.5. Blood Biomarkers

2.5.1. Peripheral T-Cell Phenotype

Surface and intracellular proteins expressed on T cells are expected to be biomarkers because ICIs target T-cell regulatory pathways. A prominent surface marker mainly expressed by CD8 effector memory T-cells is PD-1 [59]. Interestingly, PD-1 on CD8 TILs is used as a marker of tumor-reactive cells [60]. Indeed, peripheral blood PD-1+CD8 T-cells can also express neo-antigen-recognizing T-cell receptors [61].
An analysis of 29 NSCLC patients treated with PD-1 inhibitor demonstrated that 70% of patients with disease progression lacked a PD-1+CD8 T-cell response, whereas 80% of patients with a clinical response showed PD-1+CD8 T-cell responses within 4 weeks from the induction of treatment Kamphorst et al. [62].
Furthermore, CX3C chemokine receptor 1 (CX3CR1) is a receptor of the chemokine CX3CL1, which is involved in the adhesion and migration of leukocytes [63][64]. Furthermore, CX3CR1 is a marker of T-cell differentiation and is rigidly expressed on CD8+ T cells through irreversible differentiation from CX3CR1CD8+ T cells during the effector phase [64][65], which theoretically provides an advantage as a biomarker compared with transiently expressed molecules on T cells. Yamaguchi et al. reported that an increase in the frequency of the CX3CR1+ subset in circulating CD8+ T cells early after ICI therapy correlated with response and survival in 36 NSCLC patients [66].
The CD62Llow T-cell subpopulation in tumor-draining lymph nodes contains antitumor T cells and mediates potent antitumor activity when intravenously transferred [67][68]. Kgamu et al. reported that patients who responded to ICIs had a significantly higher ratio of effector CD62Llow CD4+ T cells in their peripheral blood before treatment, and that a decreased CD62Llow CD4+ T-cell ratio after ICI treatment resulted in resistance, with long-term survivors maintaining a high proportion of CD62Llow CD4+ T-cells [69].

2.5.2. Neutrophil-to-Lymphocyte Ratio

The neutrophil-to-lymphocyte ratio (NLR) is a surrogate marker of general host immune response to various stress stimuli. The systemic inflammatory response of cancer prompts neutrophil infiltration, resulting in the secretion of interleukin-2 (IL-2), interleukin-6 (IL-6), interleukin-10 (IL-10), tumor necrosis factor α (TNF-α), and vascular endothelial growth factor (VEGF) [70]. The activation and intratumoral invasion of lymphocytes are thought to be necessary for the antitumor activity of ICIs [71], while TNF-α and IL-10 cause lymphocyte dysfunction and a decrease in lymphocyte numbers [72][73].

2.5.3. Interferon-Gamma

Interferon-gamma (IFN-γ) is a cytokine that plays a role in innate and adaptive immunity, and is produced predominantly by T cells and NK cells for innate immunity but also by CD4+ and CD8+ T cells for adaptive immunity [74]. In tumors, TILs are the primary source of IFN-γ [75]. IFN-γ engages JAK/STAT signaling in the tumor cell, which induces MHC class I expression, accumulates effector cells, and promotes a loss of the suppressive activity of T-regs. Moreover, the deficiency of INF-γ inhibits effective innate and adaptive antitumor immunity [76][77].
Some reports suggested that a high-INF-γ level is related to the efficacy of ICIs. A study of durvalumab for previously treated NSCLC patients demonstrated that patients with a high pre-treatment IFN-γ signature (high levels of IFN-γ, LAG3, CXCL9, and PD-L1 mRNA expression) had higher ORR, PFS, and OS [78]. An analysis of 17 NSCLC patients treated with nivolumab showed a trend of a more prolonged OS in those with high INF-γ expression compared with those with low INF-γ expression [79]. In the POPLAR study evaluating atezolizumab in NSCLC patients, a high T-effector-IFNγ-associated gene expression status was correlated with prolonged OS [80].

2.5.4. Interleukin-8

As is known, interleukin-8 (IL-8) is a member of the CXC chemokine family and was initially identified as a chemotactic factor for neutrophils [81]. Furthermore, IL-8 is secreted by malignant cells and tumor stroma cells across many different tumor types [82]. Moreover, IL-8 directly affects endothelial cells, malignant cells, and cancer stem cells, and indirectly affects attracting and modulating tumor-associated myeloid cells [83][84].
Sanmamed et al. reported early increases in serum IL-8 levels as a predictor of poor outcome in small retrospective cohorts of patients with advanced melanoma or NSCLC who received ICIs [85]. A retrospective analysis demonstrated that high levels of IL-8 at the initiation of ICIs led to a poorer OS across renal cell carcinoma, melanoma, NSCLC, and urothelial cancer [86]. However, a poorer OS was also observed for other factors aside from ICI treatment, suggesting that IL-8 may also be a prognostic marker rather than a predictive biomarker of ICI treatment.

2.5.5. Blood/Tissue Composite Biomarker

Nebet et al. demonstrated that pre-treatment circulating tumor DNA (ctDNA) and peripheral CD8 T cell levels are independently associated with the durable clinical benefit of ICIs, and developed the DIREct-Pre (durable immunotherapy response estimation by immune profiling and ctDNA pre-treatment) score system, combining tumor PD-L1 expression with pre-treatment ctDNA and circulating immune cell profiling [87]. Patients with higher DIREct-Pre scores had significantly longer PFS with ICIs [87].

References

  1. World Health Organization. Cancer. Available online: https://gco.iarc.fr/today/fact-sheets-cancers (accessed on 15 December 2021).
  2. American Cancer Society. Non-Small Cell Lung Cancer. Available online: https://www.cancer.org/cancer/lung-cancer.html (accessed on 15 December 2021).
  3. Reck, M.; Rodríguez–Abreu, D.; Robinson, A.G.; Hui, R.; Csőszi, T.; Fülöp, A.; Gottfried, M.; Peled, N.; Tafreshi, A.; Cuffe, S.; et al. Updated Analysis of KEYNOTE-024: Pembrolizumab Versus Platinum-Based Chemotherapy for Advanced Non–Small-Cell Lung Cancer With PD-L1 Tumor Proportion Score of 50% or Greater. J. Clin. Oncol. 2019, 37, 537–546.
  4. Mok, T.S.K.; Wu, Y.-L.; Kudaba, I.; Kowalski, D.M.; Cho, B.C.; Turna, H.Z.; Castro, G., Jr.; Srimuninnimit, V.; Laktionov, K.K.; Bondarenko, I.; et al. Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): A randomised, open-label, controlled, phase 3 trial. Lancet 2019, 393, 1819–1830.
  5. Herbst, R.S.; Giaccone, G.; De Marinis, F.; Reinmuth, N.; Vergnenegre, A.; Barrios, C.H.; Morise, M.; Felip, E.; Andric, Z.; Geater, S.; et al. Atezolizumab for First-Line Treatment of PD-L1–Selected Patients with NSCLC. N. Engl. J. Med. 2020, 383, 1328–1339.
  6. Hellmann, M.D.; Paz-Ares, L.; Bernabe Caro, R.; Zurawski, B.; Kim, S.-W.; Carcereny Costa, E.; Park, K.; Alexandru, A.; Lupinacci, L.; de la Mora Jimenez, E.; et al. Nivolumab plus Ipilimumab in Advanced Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2019, 381, 2020–2031.
  7. Gandhi, L.; Rodríguez-Abreu, D.; Gadgeel, S.; Esteban, E.; Felip, E.; De Angelis, F.; Domine, M.; Clingan, P.; Hochmair, M.J.; Powell, S.F.; et al. Pembrolizumab plus Chemotherapy in Metastatic Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 378, 2078–2092.
  8. Paz-Ares, L.; Vicente, D.; Tafreshi, A.; Robinson, A.; Parra, H.S.; Mazières, J.; Hermes, B.; Cicin, I.; Medgyasszay, B.; Rodríguez-Cid, J.; et al. A Randomized, Placebo-Controlled Trial of Pembrolizumab Plus Chemotherapy in Patients with Metastatic Squamous NSCLC: Protocol-Specified Final Analysis of KEYNOTE-407. J. Thorac. Oncol. 2020, 15, 1657–1669.
  9. Socinski, M.A.; Jotte, R.M.; Cappuzzo, F.; Orlandi, F.; Stroyakovskiy, D.; Nogami, N.; Rodríguez-Abreu, D.; Moro-Sibilot, D.; Thomas, C.A.; Barlesi, F.; et al. Atezolizumab for First-Line Treatment of Metastatic Nonsquamous NSCLC. N. Engl. J. Med. 2018, 378, 2288–2301.
  10. West, H.; McCleod, M.; Hussein, M.; Morabito, A.; Rittmeyer, A.; Conter, H.J.; Kopp, H.-G.; Daniel, D.; McCune, S.; Mekhail, T.; et al. Atezolizumab in combination with carboplatin plus nab-paclitaxel chemotherapy compared with chemotherapy alone as first-line treatment for metastatic non-squamous non-small-cell lung cancer (IMpower130): A multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2019, 20, 924–937.
  11. Paz-Ares, L.; Ciuleanu, T.-E.; Cobo, M.; Schenker, M.; Zurawski, B.; Menezes, J.; Richardet, E.; Bennouna, J.; Felip, E.; Juan-Vidal, O.; et al. First-line nivolumab plus ipilimumab combined with two cycles of chemotherapy in patients with non-small-cell lung cancer (CheckMate 9LA): An international, randomised, open-label, phase 3 trial. Lancet Oncol. 2021, 22, 198–211.
  12. Reck, M.; Rodríguez-Abreu, D.; Robinson, A.G.; Hui, R.; Csőszi, T.; Fülöp, A.; Gottfried, M.; Peled, N.; Tafreshi, A.; Cuffe, S.; et al. Five-Year Outcomes with Pembrolizumab Versus Chemotherapy for Metastatic Non–Small-Cell Lung Cancer With PD-L1 Tumor Proportion Score ≥ 50%. J. Clin. Oncol. 2021, 39, 2339–2349.
  13. Borghaei, H.; Gettinger, S.; Vokes, E.E.; Chow, L.Q.M.; Burgio, M.A.; Carpeno, J.D.C.; Pluzanski, A.; Arrieta, O.; Frontera, O.A.; Chiari, R.; et al. Five-Year Outcomes from the Randomized, Phase III Trials CheckMate 017 and 057: Nivolumab Versus Docetaxel in Previously Treated Non–Small-Cell Lung Cancer. J. Clin. Oncol. 2021, 39, 723–733.
  14. Kumar, V.; Chaudhary, N.; Garg, M.; Floudas, C.S.; Soni, P.; Chandra, A.B. Current diagnosis and management of immune related adverse events (irAEs) induced by immune checkpoint inhibitor therapy. Front. Pharmacol. 2017, 8, 49.
  15. Puzanov, I.; Diab, A.; Abdallah, K.; Bingham, C.O., 3rd; Brogdon, C.; Dadu, R.; Hamad, L.; Kim, S.; Lacouture, M.E.; LeBoeuf, N.R.; et al. Managing toxicities associated with immune checkpoint inhibitors: Consensus recommendations from the Society for Immunotherapy of Cancer (SITC) Toxicity Management Working Group. J. Immunother. Cancer 2017, 5, 95.
  16. Winer, A.; Bodor, J.N.; Borghaei, H. Identifying and managing the adverse effects of immune checkpoint blockade. J. Thorac. Dis. 2018, 10, S480–S489.
  17. Chae, Y.K.; Viveiros, P.; Lopes, G.; Sukhadia, B.; Sheikh, M.M.; Saravia, D.; Florou, V.; Sokol, E.S.; Frampton, G.M.; Chalmers, Z.R.; et al. Clinical and Immunological Implications of Frameshift Mutations in Lung Cancer. J. Thorac. Oncol. 2019, 14, 1807–1817.
  18. Łuksza, M.; Riaz, N.; Makarov, V.; Balachandran, V.P.; Hellmann, M.D.; Solovyov, A.; Rizvi, N.A.; Merghoub, T.; Levine, A.J.; Chan, T.A.; et al. A neoantigen fitness model predicts tumour response to checkpoint blockade immunotherapy. Nature 2017, 551, 517–520.
  19. Matsushita, H.; Vesely, M.D.; Koboldt, D.C.; Rickert, C.G.; Uppaluri, R.; Magrini, V.J.; Arthur, C.D.; White, J.M.; Chen, Y.S.; Shea, L.K.; et al. Cancer exome analysis reveals a T-cell-dependent mechanism of cancer immunoediting. Nature 2012, 482, 400–404.
  20. Ott, P.A.; Hu, Z.; Keskin, D.B.; Shukla, S.A.; Sun, J.; Bozym, D.J.; Zhang, W.; Luoma, A.; Giobbie-Hurder, A.; Peter, L.; et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature 2017, 547, 217–221, Erratum in Nature 2018, 555, 402.
  21. Rizvi, N.A.; Cho, B.C.; Reinmuth, N.; Lee, K.H.; Luft, A.; Ahn, M.-J.; Van Den Heuvel, M.M.; Cobo, M.; Vicente, D.; Smolin, A.; et al. Durvalumab with or Without Tremelimumab vs Standard Chemotherapy in First-line Treatment of Metastatic Non-Small Cell Lung Cancer: The MYSTIC Phase 3 Randomized Clinical Trial. JAMA Oncol. 2020, 6, 661–674.
  22. Govindan, R.; Ding, L.; Griffith, M.; Subramanian, J.; Dees, N.D.; Kanchi, K.L.; Maher, C.A.; Fulton, R.; Fulton, L.; Wallis, J.; et al. Genomic Landscape of Non-Small Cell Lung Cancer in Smokers and Never-Smokers. Cell 2012, 150, 1121–1134.
  23. Ramalingam, S.S.; Hellmann, M.D.; Awad, M.M.; Borghaei, H.; Gainor, J.; Brahmer, J.; Spigel, D.R.; Reck, M.; O’Byrne, K.J.; Paz-Ares, L.; et al. Abstract CT078: Tumor mutational burden (TMB) as a biomarker for clinical benefit from dual immune checkpoint blockade with nivolumab (nivo) + ipilimumab (ipi) in first-line (1L) non-small cell lung cancer (NSCLC): Identification of TMB cutoff from CheckMate 568. Cancer Res. 2018, 78, CT078.
  24. Hellmann, M.D.; Ciuleanu, T.-E.; Pluzanski, A.; Lee, J.S.; Otterson, G.A.; Audigier-Valette, C.; Minenza, E.; Linardou, H.; Burgers, S.; Salman, P.; et al. Nivolumab plus Ipilimumab in Lung Cancer with a High Tumor Mutational Burden. N. Engl. J. Med. 2018, 378, 2093–2104.
  25. 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.
  26. Merino, D.M.; McShane, L.M.; Fabrizio, D.; Funari, V.; Chen, S.J.; White, J.R.; Wenz, P.; Baden, J.; Barrett, J.C.; Chaudhary, R.; et al. Establishing guidelines to harmonize tumor mutational burden (TMB): In silico assessment of variation in TMB quantification across diagnostic platforms: Phase I of the Friends of Cancer Research TMB Harmonization Project. J. Immunother. Cancer 2020, 8, e000147.
  27. Chalmers, Z.R.; Connelly, C.F.; Fabrizio, D.; Gay, L.; Ali, S.M.; Ennis, R.; Schrock, A.; Campbell, B.; Shlien, A.; Chmielecki, J.; et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017, 9, 34.
  28. Rizvi, H.; Sanchez-Vega, F.; La, K.; Chatila, W.; Jonsson, P.; Halpenny, D.; Plodkowski, A.; Long, N.; Sauter, J.L.; Rekhtman, N.; et al. Molecular Determinants of Response to Anti–Programmed Cell Death (PD)-1 and Anti–Programmed Death-Ligand 1 (PD-L1) Blockade in Patients with Non–Small-Cell Lung Cancer Profiled with Targeted Next-Generation Sequencing. J. Clin. Oncol. 2018, 36, 633–641.
  29. Marabelle, A.; Fakih, M.; Lopez, J.; Shah, M.; Shapira-Frommer, R.; Nakagawa, K.; Chung, H.; Kindler, H.; Lopez-Martin, J.; Miller, A. Association of tumor mutational burden with outcomes in patients with select advanced solid tumors treated with pembrolizumab in KEYNOTE-158. ESMO 2019 congress. Ann. Oncol. 2019, 30, v475–v532.
  30. Garassino, M.; Rodriguez-Abreu, D.; Gadgeel, S.; Esteban, E.; Felip, E.; Speranza, G.; Reck, M.; Hui, R.; Boyer, M.; Cristescu, R.; et al. Evaluation of TMB in KEYNOTE-189: Pembrolizumab plus chemotherapy vs. placebo plus chemotherapy for nonsquamous NSCLC. J. Thorac. Oncol. 2019, 14, S216–S217.
  31. Langer, C.; Gadgeel, S.; Borghaei, H.; Patnaik, A.; Powell, S.; Gentzler, R.; Yang, J.; Gubens, M.; Sequist, L.; Awad, M.; et al. OA04.05 KEYNOTE-021: TMB and Outcomes for Carboplatin and Pemetrexed with or Without Pembrolizumab for Nonsquamous NSCLC. J. Thorac. Oncol. 2019, 14, S216.
  32. Anagnostou, V.; Niknafs, N.; Marrone, K.; Bruhm, D.C.; White, J.R.; Naidoo, J.; Hummelink, K.; Monkhorst, K.; Lalezari, F.; Lanis, M.; et al. Multimodal genomic features predict outcome of immune checkpoint blockade in non-small-cell lung cancer. Nat. Cancer 2020, 1, 99–111.
  33. McGranahan, N.; Furness, A.J.S.; Rosenthal, R.; Ramskov, S.; Lyngaa, R.; Saini, S.K.; Jamal-Hanjani, M.; Wilson, G.A.; Birkbak, N.J.; Hiley, C.T.; et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016, 351, 1463–1469.
  34. Modrich, P.; Lahue, R. Mismatch repair in replication fidelity, genetic recombination, and cancer biology. Annu. Rev. Biochem. 1996, 65, 101–133.
  35. Begum, R.; Martin, S.A. Targeting Mismatch Repair defects: A novel strategy for personalized cancer treatment. DNA Repair 2016, 38, 135–139.
  36. Hause, R.J.; Pritchard, C.C.; Shendure, J.; Salipante, S.J. Classification and characterization of microsatellite instability across 18 cancer types. Nat. Med. 2016, 22, 1342–1350.
  37. Le, D.T.; Durham, J.N.; Smith, K.N.; Wang, H.; Bartlett, B.R.; Aulakh, L.K.; Lu, S.; Kemberling, H.; Wilt, C.; Luber, B.S.; et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017, 357, 409–413.
  38. Bonneville, R.; Krook, M.A.; Kautto, E.A.; Miya, J.; Wing, M.R.; Chen, H.-Z.; Reeser, J.W.; Yu, L.; Roychowdhury, S. Landscape of Microsatellite Instability Across 39 Cancer Types. JCO Precis. Oncol. 2017, 1, 1–15.
  39. Quail, D.F.; Joyce, J.A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 2013, 19, 1423–1437.
  40. DeNardo, D.G.; Andreu, P.; Coussens, L.M. Interactions between lymphocytes and myeloid cells regulate pro- versus anti-tumor immunity. Cancer Metastasis Rev. 2010, 29, 309–316.
  41. Li, F.; Li, C.; Cai, X.; Xie, Z.; Zhou, L.; Cheng, B.; Zhong, R.; Xiong, S.; Li, J.; Chen, Z.; et al. The association between CD8+ tumor-infiltrating lymphocytes and the clinical outcome of cancer immunotherapy: A systematic review and meta-analysis. eClinicalMedicine 2021, 41, 101134.
  42. Fumet, J.-D.; Richard, C.; Ledys, F.; Klopfenstein, Q.; Joubert, P.; Routy, B.; Truntzer, C.; Gagné, A.; Hamel, M.-A.; Guimaraes, C.F.; et al. Prognostic and predictive role of CD8 and PD-L1 determination in lung tumor tissue of patients under anti-PD-1 therapy. Br. J. Cancer 2018, 119, 950–960.
  43. Niemeijer, A.-L.N.; Sahba, S.; Smit, E.F.; Lissenberg-Witte, B.I.; de Langen, A.J.; Thunnissen, E. Association of tumour and stroma PD-1, PD-L1, CD3, CD4 and CD8 expression with DCB and OS to nivolumab treatment in NSCLC patients pre-treated with chemotherapy. Br. J. Cancer 2020, 123, 392–402.
  44. Hu-Lieskovan, S.; Lisberg, A.; Zaretsky, J.M.; Grogan, T.R.; Rizvi, H.; Wells, D.K.; Carroll, J.; Cummings, A.; Madrigal, J.; Jones, B.; et al. Tumor Characteristics Associated with Benefit from Pembrolizumab in Advanced Non–Small Cell Lung Cancer. Clin. Cancer Res. 2019, 25, 5061–5068.
  45. Hashemi, S.; Fransen, M.; Niemeijer, A.; Ben Taleb, N.; Houda, I.; Veltman, J.; Becker-Commissaris, A.; Daniels, H.; Crombag, L.; Radonic, T.; et al. Surprising impact of stromal TIL’s on immunotherapy efficacy in a real-world lung cancer study. Lung Cancer 2021, 153, 81–89.
  46. Althammer, S.; Tan, T.H.; Spitzmüller, A.; Rognoni, L.; Wiestler, T.; Herz, T.; Widmaier, M.; Rebelatto, M.C.; Kaplon, H.; Damotte, D.; et al. Automated image analysis of NSCLC biopsies to predict response to anti-PD-L1 therapy. J. Immunother. Cancer 2019, 7, 121.
  47. 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.
  48. Shirasawa, M.; Yoshida, T.; Shimoda, Y.; Takayanagi, D.; Shiraishi, K.; Kubo, T.; Mitani, S.; Matsumoto, Y.; Masuda, K.; Shinno, Y.; et al. Differential Immune-Related Microenvironment Determines Programmed Cell Death Protein-1/Programmed Death-Ligand 1 Blockade Efficacy in Patients With Advanced NSCLC. J. Thorac. Oncol. 2021, 16, 2078–2090.
  49. Kumagai, S.; Togashi, Y.; Kamada, T.; Sugiyama, E.; Nishinakamura, H.; Takeuchi, Y.; Vitaly, K.; Itahashi, K.; Maeda, Y.; Matsui, S.; et al. The PD-1 expression balance between effector and regulatory T cells predicts the clinical efficacy of PD-1 blockade therapies. Nat. Immunol. 2020, 21, 1346–1358.
  50. Tumeh, P.C.; Harview, C.L.; Yearley, J.H.; Shintaku, I.P.; Taylor, E.J.M.; Robert, L.; Chmielowski, B.; Spasic, M.; Henry, G.; Ciobanu, V.; et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014, 515, 568–571.
  51. Kiepiela, P.; Leslie, A.; Honeyborne, I.; Ramduth, D.; Thobakgale-Tshabalala, C.; Chetty, S.; Rathnavalu, P.; Moore, C.C.; Pfafferott, K.J.; Hilton, L.; et al. Dominant influence of HLA-B in mediating the potential co-evolution of HIV and HLA. Nature 2004, 432, 769–775.
  52. Chowell, D.; Krishna, S.; Becker, P.D.; Cocita, C.; Shu, J.; Tan, X.; Greenberg, P.D.; Klavinskis, L.S.; Blattman, J.N.; Anderson, K.S. TCR contact residue hydrophobicity is a hallmark of immunogenic CD8 + T cell epitopes. Proc. Natl. Acad. Sci. USA 2015, 112, E1754–E1762.
  53. Parham, P.; Ohta, T. Population Biology of Antigen Presentation by MHC Class I Molecules. Science 1996, 272, 67–74.
  54. Naranbhai, V.; Viard, M.; Dean, M.; Groha, S.; Braun, D.A.; Labaki, C.; Shukla, S.A.; Yuki, Y.; Shah, P.; Chin, K.; et al. HLA-A*03 and response to immune checkpoint blockade in cancer: An epidemiological biomarker study. Lancet Oncol. 2022, 23, 172–184.
  55. Chowell, D.; Morris, L.G.T.; Grigg, C.M.; Weber, J.K.; Samstein, R.M.; Makarov, V.; Kuo, F.; Kendall, S.M.; Requena, D.; Riaz, N.; et al. Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science 2018, 359, 582–587.
  56. Abed, A.; Calapre, L.; Lo, J.; Correia, S.; Bowyer, S.; Chopra, A.; Watson, M.; Khattak, M.A.; Millward, M.; Gray, E.S. Prognostic value of HLA-I homozygosity in patients with non-small cell lung cancer treated with single agent immunotherapy. J. Immunother. Cancer 2020, 8, e001620.
  57. Negrao, M.; Lam, V.K.; Reuben, A.; Rubin, M.L.; Landry, L.L.; Roarty, E.B.; Rinsurongkawong, W.; Lewis, J.; Roth, J.A.; Swisher, S.G.; et al. PD-L1 Expression, Tumor Mutational Burden, and Cancer Gene Mutations Are Stronger Predictors of Benefit from Immune Checkpoint Blockade than HLA Class I Genotype in Non–Small Cell Lung Cancer. J. Thorac. Oncol. 2019, 14, 1021–1031.
  58. Schaafsma, E.; Fugle, C.M.; Wang, X.; Cheng, C. Pan-cancer association of HLA gene expression with cancer prognosis and immunotherapy efficacy. Br. J. Cancer 2021, 125, 422–432.
  59. Baitsch, L.; Legat, A.; Barba, L.; Marraco, S.A.F.; Rivals, J.-P.; Baumgaertner, P.; Christiansen-Jucht, C.; Bouzourene, H.; Rimoldi, D.; Pircher, H.; et al. Extended Co-Expression of Inhibitory Receptors by Human CD8 T-Cells Depending on Differentiation, Antigen-Specificity and Anatomical Localization. PLoS ONE 2012, 7, e30852.
  60. Gros, A.; Robbins, P.F.; Yao, X.; Li, Y.F.; Turcotte, S.; Tran, E.; Wunderlich, J.R.; Mixon, A.; Farid, S.; Dudley, M.E.; et al. PD-1 identifies the patient-specific CD8+ tumor-reactive repertoire infiltrating human tumors. J. Clin. Investig. 2014, 124, 2246–2259.
  61. Gros, A.; Parkhurst, M.R.; Tran, E.; Pasetto, A.; Robbins, P.F.; Ilyas, S.; Prickett, T.D.; Gartner, J.J.; Crystal, J.S.; Roberts, I.M.; et al. Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients. Nat. Med. 2016, 22, 433–438.
  62. 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.
  63. Böttcher, J.; Beyer, M.; Meissner, F.; Abdullah, Z.; Sander, J.; Höchst, B.; Eickhoff, S.; Rieckmann, J.C.; Russo, C.; Bauer, T.; et al. Functional classification of memory CD8+ T cells by CX3CR1 expression. Nat. Commun. 2015, 6, 8306.
  64. Gerlach, C.; Moseman, E.A.; Loughhead, S.M.; Alvarez, D.; Zwijnenburg, A.J.; Waanders, L.; Garg, R.; de la Torre, J.C.; von Andrian, U.H. The Chemokine Receptor CX3CR1 Defines Three Antigen-Experienced CD8 T Cell Subsets with Distinct Roles in Immune Surveillance and Homeostasis. Immunity 2016, 45, 1270–1284.
  65. Gordon, C.L.; Lee, L.N.; Swadling, L.; Hutchings, C.; Zinser, M.; Highton, A.J.; Capone, S.; Folgori, A.; Barnes, E.; Klenerman, P. Induction and Maintenance of CX3CR1-Intermediate Peripheral Memory CD8+ T Cells by Persistent Viruses and Vaccines. Cell Rep. 2018, 23, 768–782.
  66. Yamauchi, T.; Hoki, T.; Oba, T.; Jain, V.; Chen, H.; Attwood, K.; Battaglia, S.; George, S.; Chatta, G.; Puzanov, I.; et al. T-cell CX3CR1 expression as a dynamic blood-based biomarker of response to immune checkpoint inhibitors. Nat. Commun. 2021, 12, 1402.
  67. Kagamu, H.; Touhalisky, J.E.; Plautz, G.E.; Krauss, J.C.; Shu, S. Isolation based on L-selectin expression of immune effector T cells derived from tumor-draining lymph nodes. Cancer Res. 1996, 56, 4338–4342.
  68. Kagamu, H.; Shu, S. Purification of L-selectinlow cells promotes the generation of highly potent CD4 antitumor effector T lymphocytes. J. Immunol. 1998, 160, 3444–3452.
  69. Kagamu, H.; Kitano, S.; Yamaguchi, O.; Yoshimura, K.; Horimoto, K.; Kitazawa, M.; Fukui, K.; Shiono, A.; Mouri, A.; Nishihara, F.; et al. CD4+ T-cell Immunity in the Peripheral Blood Correlates with Response to Anti-PD-1 Therapy. Cancer Immunol. Res. 2020, 8, 334–344.
  70. Yang, J.-J.; Hu, Z.-G.; Shi, W.-X.; Deng, T.; He, S.-Q.; Yuan, S.-G. Prognostic significance of neutrophil to lymphocyte ratio in pancreatic cancer: A meta-analysis. World J. Gastroenterol. 2015, 21, 2807–2815.
  71. Petrie, H.T.; Klassen, L.W.; Kay, H.D. Inhibition of human cytotoxic T lymphocyte activity in vitro by autologous peripheral blood granulocytes. J. Immunol. 1985, 134, 230–234.
  72. Bellone, G.; Turletti, A.; Artusio, E.; Mareschi, K.; Carbone, A.; Tibaudi, D.; Robecchi, A.; Emanuelli, G.; Rodeck, U. Tumor-Associated Transforming Growth Factor-β and Interleukin-10 Contribute to a Systemic Th2 Immune Phenotype in Pancreatic Carcinoma Patients. Am. J. Pathol. 1999, 155, 537–547.
  73. Salazar-Onfray, F.; López, M.N.; Mendoza-Naranjo, A. Paradoxical effects of cytokines in tumor immune surveillance and tumor immune escape. Cytokine Growth Factor Rev. 2007, 18, 171–182.
  74. Critchley-Thorne, R.J.; Simons, D.L.; Yan, N.; Miyahira, A.K.; Dirbas, F.M.; Johnson, D.L.; Swetter, S.M.; Carlson, R.W.; Fisher, G.A.; Koong, A.; et al. Impaired interferon signaling is a common immune defect in human cancer. Proc. Natl. Acad. Sci. USA 2009, 106, 9010–9015.
  75. Ni, L.; Lu, J. Interferon gamma in cancer immunotherapy. Cancer Med. 2018, 7, 4509–4516.
  76. Zaretsky, J.M.; Garcia-Diaz, A.; Shin, D.S.; Escuin-Ordinas, H.; Hugo, W.; Hu-Lieskovan, S.; Torrejon, D.Y.; Abril-Rodriguez, G.; Sandoval, S.; Barthly, L.; et al. Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. N. Engl. J. Med. 2016, 375, 819–829.
  77. Spranger, S.; Spaapen, R.M.; Zha, Y.; Williams, J.; Meng, Y.; Ha, T.T.; Gajewski, T.F. Up-regulation of PD-L1, IDO, and Tregs in the melanoma tumor microenvironment is driven by CD8+ T cells. Sci. Transl. Med. 2013, 5, 200ra116.
  78. Higgs, B.W.; Morehouse, C.A.; Streicher, K.; Brohawn, P.Z.; Steele, K.; Rebelatto, M.; Pilataxi, F.; Bais, C.; Shi, L.; Jin, X.; et al. Abstract 1773: A baseline IFNG gene expression signature correlates with clinical outcomes in durvalumab-treated advanced NSCLC cancer patients. Cancer Res. 2017, 77, 1773.
  79. Karachaliou, N.; Gonzalez-Cao, M.; Crespo, G.; Drozdowskyj, A.; Aldeguer, E.; Gimenez-Capitan, A.; Teixido, C.; Molina-Vila, M.A.; Viteri, S.; de los Llanos Gil, M.; et al. Interferon gamma, an important marker of response to immune checkpoint blockade in non-small cell lung cancer and melanoma patients. Ther. Adv. Med. Oncol. 2018, 10, 1758834017749748.
  80. Fehrenbacher, L.; Spira, A.; Ballinger, M.; Kowanetz, M.; Vansteenkiste, J.; Mazieres, J.; Park, K.; Smith, D.; Artal-Cortes, A.; Lewanski, C.; et al. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): A multicentre, open-label, phase 2 randomised controlled trial. Lancet 2016, 387, 1837–1846.
  81. Walz, A.; Peveri, P.; Aschauer, H.; Baggiolini, M. Purification and amino acid sequencing of NAF, a novel neutrophil-activating factor produced by monocytes. Biochem. Biophys. Res. Commun. 1987, 149, 755–761.
  82. Zarogoulidis, P.; Katsikogianni, F.; Tsiouda, T.; Sakkas, A.; Katsikogiannis, N.; Zarogoulidis, K. Interleukin-8 and interleukin-17 for cancer. Cancer Investig. 2014, 32, 197–205.
  83. Yuan, A.; Chen, J.J.W.; Yao, P.-L.; Yang, P.-C. The role of interleukin-8 in cancer cells and microenvironment interaction. Front. Biosci. 2005, 10, 853–865.
  84. Mantovani, A.; Germano, G.; Marchesi, F.; Locatelli, M.; Biswas, S.K. Cancer-promoting tumor-associated macrophages: New vistas and open questions. Eur. J. Immunol. 2011, 41, 2522–2525.
  85. Sanmamed, M.F.; Perez-Gracia, J.L.; Schalper, K.A.; Fusco, J.P.; Gonzalez, A.; Rodriguez-Ruiz, M.E.; Oñate, C.; Perez, G.; Alfaro, C.; Martín-Algarra, S.; et al. Changes in serum interleukin-8 (IL-8) levels reflect and predict response to anti-PD-1 treatment in melanoma and non-small-cell lung cancer patients. Ann. Oncol. 2017, 28, 1988–1995.
  86. Schalper, K.A.; Carleton, M.; Zhou, M.; Chen, T.; Feng, Y.; Huang, S.-P.; Walsh, A.M.; Baxi, V.; Pandya, D.; Baradet, T.; et al. Elevated serum interleukin-8 is associated with enhanced intratumor neutrophils and reduced clinical benefit of immune-checkpoint inhibitors. Nat. Med. 2020, 26, 688–692.
  87. Nabet, B.Y.; Esfahani, M.S.; Moding, E.J.; Hamilton, E.G.; Chabon, J.J.; Rizvi, H.; Steen, C.B.; Chaudhuri, A.A.; Liu, C.L.; Hui, A.B.; et al. Noninvasive Early Identification of Therapeutic Benefit from Immune Checkpoint Inhibition. Cell 2020, 183, 363–376.e13.
More
Information
Subjects: Oncology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 233
Revisions: 2 times (View History)
Update Date: 01 Apr 2022
1000/1000