#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Programmed death-ligand 1 expression in non-small cell lung carcinoma – mechanism of regulation, association with other markers, and therapeutic implication


Expresia ligandu 1 programovanej smrti v nemalobunkovom karcinóme pľúc – mechanizmus regulácie, asociácia s ostatnými markermi a terapeutické využitie

Východiská: Inhibítory imunitných kontrolných bodov (ICI) blokujúce signálnu dráhu proteínu 1 programovanej smrti (PD-1), dramaticky zlepšili prežívanie pacientov s pokročilým nemalobunkovým karcinómom pľúc (NSCLC). Imunohistochemická analýza expresie ligandu 1 programovanej smrti (PD-L1) je toho času najviac využívaným a klinicky validovaným bio­markerom predikujúcim efektívnosť ICI u pacientov s NSCLC, ale sám o sebe predstavuje nedokonalý nástroj. Signálna dráha PD-1 je poprepájaná s početnými celulárnymi ako aj molekulárnymi faktormi prítomnými v nádorovom mikroprostredí (TME) v NSCLC. Celulárne faktory, ktoré sa podieľajú na regulácii expresie PD-L1 v NSCLC sú pripisované aktivite nádor infiltrujúcich lymfocytov a s nádorom asociovanými fibroblastmi. Vnútorné molekulárne faktory, ktoré majú vplyv na úroveň expresie PD-L1 v NSCLC, sú asociované s prítomnosťou onkogénnych driver mutácií v génoch receptora epidermálneho rastového faktora a v homológu virového onkogénu Kirsten rat sarcoma a s translokáciami vedúcimi k prestavbe kinázy anaplastického lymfómu. Okrem toho, na úroveň expresie PD-L1 v NSCLC môže mať vplyv aj stimulácia hypoxických signálnych dráh a aktivácia transformujúceho rastového faktora beta 1. Hlbšie pochopenie zložitých mechanizmov regulujúcich expresiu PD-L1 je nevyhnutné, aby bolo v budúcnosti možné ušiť na mieru terapiu s použitím ICI u pacientov s pokročilým NSCLC. Cieľ: V predkladanom prehľadovom článku prezentujeme súhrn kľúčových faktorov podieľajúcich sa na regulácii expresie PD-L1 v rámci TME v NSCLC, ktoré sú a potenciálne môžu byť využívané za účelom zlepšenia účinnosti imunoterapie, ktorá blokuje signálnu dráhu PD-1.

Klíčová slova:

nemalobunkový karcinóm pľúc – tumor infiltrujúce lymfocyty – inhibítory imunitných kontrolných bodov – proteínu 1 programovanej smrti – ligand 1 programovanej bunkovej smrti – epitelovo mezenchýmový prechod – hypoxiou indukovateľný faktor-1α


Authors: V. Tancoš 1;  A. Blichárová 1;  L. Plank 2,3
Authors place of work: Ústav patológie UPJŠ LF a UNLP, Slovenská republika 1;  Ústav patologickej anatómie Jesseniovej lekárskej fakulty Univerzity Komenského a Univerzitnej nemocnice v Martine, Slovenská republika 2;  Martinské bio ptické centrum, s. r. o. v Martin, Slovenská republika 3
Published in the journal: Klin Onkol 2022; 35(5): 372-376
Category: Přehled
doi: https://doi.org/10.48095/ccko2022372

Summary

Background: Immune checkpoint inhibitors (ICI) targeting the programmed cell death protein 1 (PD-1) signaling pathway have dramatically improved the clinical outcomes of oncological patients having advanced non-small cell lung carcinoma (NSCLC). The immunohistochemical analysis of programmed death-ligand 1 (PD-L1) expression remains the most widely used and clinically validated bio­marker predicting efficacy of ICI in NSCLC patients, but it represents in isolation an imperfect tool. The PD-1 axis is intricately coupled with numerous cellular and molecular factors within the tumor microenvironment (TME) of NSCLC. Cellular factors implicated in the regulation process of PD-L1 expression in NSCLC are related to the activity of tumor infiltrating lymphocytes and cancer associated fibroblasts. Intrinsic molecular factors which affect the level of PD-L1 expression are associated with the presence of oncogenic driver mutations in the Kirsten rat sarcoma viral oncogene homolog and epidermal growth factor receptor genes and to rearrangements in the anaplastic lymphoma kinase. Furthermore, activation of hypoxic signaling pathways and the transforming growth factor beta 1 axis can have an impact on the level of PD-L1 expression in NSCLC. A deeper understanding of the complex mechanisms regulating PD-L1 expression is necessary to tailor the treatment with ICI in patients with advanced NSCLC. Purpose: In this review, we present an overview of key factors underlying the regulation of PD-L1 expression within the TME of NSCLC, which are, and potentially can be, exploited to improve the outcomes of immunotherapy targeting the PD-1 axis.

Keywords:

tumor infiltrating lymphocytes – programmed death-ligand 1 – non-small cell lung carcinoma – immune checkpoint inhibitors – programmed cell death protein 1 – epithelial to mesenchymal transition – hypoxia -inducible factor-1α

Introduction

Programmed cell death protein1 (PD-1) is a type I transmembrane protein which is expressed on the surface of activated immune cells [1]. PD-1 is acting mainly as a receptor which engages into interaction with programmed death-ligand 1 (PD-L1; encoded by CD274, present on locus 9p24.1) and, to a lesser degree, with PD-L2 [2]. PD-L1 is the major ligand which activates the PD-1 signaling pathway [3]. PD-L1 is expressed on the surface of normal cells, where it safeguards tolerance and hampers exaggerated and potentially harmful immune responses in places where inflammation rages [4]. However, the process of clonal expansion in cancer leads to the selection of malignant cells with the ability to aberrantly express PD-L1, which en­ables them to evade elimination by tumor-specific immune responses [5]. Immune checkpoint inhibitors (ICI), which aim to block the interaction of PD-1 with its ligand PD-L1, heralded a new era in oncological therapy and significantly improved overall survival as well as the quality of life of patients with locally advanced or metastatic non-small cell lung carcinoma (NSCLC) [6–9]. In clinical studies, survival benefit and efficacy of ICI has been linked to PD-L1 positivity in NSCLC [8,10,11]. Expression of PD-L1 on tumor cells determined immunohistochemically remains the mainstay predictive factor for indication of immunotherapy in NSCLC patients [12], but it represents an imperfect marker [13]. PD-L1 negativity does not always preclude effectiveness and survival benefit from immunotherapy, and some highly PD-L1 positive NSCLCs have been reported as being irresponsive to ICI treatment [14]. These ostensible limitations can be attributed to the intricating nature of the PD-1 axis, which may be, eventually, surpassed after clarification of the numerous interconnections between PD-L1 regulation and other factors and signaling pathways present in the framework of the tumor microenvironment (TME) [15].

In this review, we present an overview of key factors underlying the regulation of PD-L1 expression in the TME of NSCLC, which are, and potentially can be, exploited to improve outcomes of immunotherapy targeting the PD-1 axis.

Inflammatory-driven  PD-L1 expression

Under normal physiological conditions, expression of PD-L1 is up-regulated in cells of peripheral tissues as a response to prolonged and exaggerated action of pro-inflammatory cytokines elaborated by activated immune cells [16]. Malignant cells of NSCLC are able adopt this extrinsic way of PD-L1 expression [17].

Out of the multitude of pro-inflammatory cytokines secreted by immune cells, interferon- g (IFN- g) is considered the most robust external factor, which leads to substantial increase in the level of PD-L1 expression in tumor cells [18]. The IFN- g-mediated up-regulation of PD-L1 is conducted mainly through the activation of the Janus kinase (JAK) / signal transducer and activator of transcription (STAT) / interferon responsive factor 1 (IRF1) signaling pathways [19].

Besides IFN- g, tumor necrosis factor a (TNF-a), and several interleukins (IL-6, IL-10, and IL-27) act synergistically with IFN- g and lead to induction of PD-L1 expression in the TME [20–24]. TNF-a activates transcription of CD274 through stimulation of the nuclear factor kap­pa-B (NF-kB) signaling pathway [20]. IL-6 launched the STAT3 / c-MYC / miR-25-3p axis with a subsequent decrease in the type O protein tyrosine phosphatase receptor (PTPRO) [21]. Down-regulation of PTPRO deregulated the activation of the JAK2 – STAT1 / STAT3 signaling, which eventually led to increased PD-L1 expres- sion [21]. Multiple pathways are implicated in IL-6-driven PD-L1 expression in NSCLC, especially the activation of MEK – ERK [22]. IL-10 causes a significant up-regulation of PD-L1 in tumor-associated macrophages [23]. IL-27 promoted the phosphorylation of tyrosine residues in STAT1 and STAT3, which caused an in­crease in transcription of CD274 and up-regulation of PD-L1 in several types of human malignancies, including lung cancer [24].

Overall, many pro-inflammatory cytokines are embroiled in the process of PD-L1 expression within the TME, which suggest new therapeutic strategies in the future [25]. But because these external factors are elaborated by activated immune cells, a TME with inflammatory characteristics may be a requirement when aiming to interfere with PD-L1 expression in tumor cells by inflammation-modulating drugs.

Cellular factors in the TME associated with PD-L1 expression

An inflammatory TME is characterized by the presence of a cellular infiltrate rich in lymphocytes, which aim to eliminate malignant-transformed cells, but usually fail to fulfil their quest because of the ability of tumors to make use of the PD-1 axis [12]. Quantitative measurements of immune cells occupying the TME as well as evaluation of their immune products are intensely studied as possible predictors of clinical efficacy of ICI therapy, potentially complementing PD-L1 immunohistochemistry [26]. Assessment of the density of T-cell present within tumors has gained the most attention so far as being a clinically relevant and laboratory relatively easily accessible predictive factor besides PD-L1 [27]. Abundance of tumor infiltrating lymphocytes (TIL) present at baseline evaluation has been shown to be associated with higher efficacy of ICI in a multitude of malignancies [26,28–29]. Inflammatory signatures indicative of competent and functioning antitumor immunity are also of predictive value [30]. Better clinical responses were observed in oncological patients having their tumors infiltrated by immune cells, characterized by an intracellular content rich in IFN- g-related mRNA [30,31]. In patients with locally advanced or metastatic NSCLC, the presence of a high amount of CD8+ TIL within the TME and/or a higher intracel­lular content of CD8A mRNA transcripts determined immunohistochemically have been associated with better progression free survival, when treatment with ICI was applied [32]. The predictive value of these factors was significantly amplified when combined with the PD-L1 immunohistochemical analysis at the protein and/or mRNA level [32]. It was demonstrated in another study, that high levels of CD3+ TIL in slides evaluated using multiplex quantitative immunofluorescence significantly correlated with a better durable clinical benefit and overall survival in NSCLC patients treated with ICI [33].

These results suggest that the integration of all these markers may provide a more robust framework for developing an effective therapeutic algorithm.

PD-L1 expression  in oncogenic-driven NSCLC

Various genetic alterations and epigenetic modifiers can lead to constitutive expression of PD-L1 in tumor cells of lung cancer [25]. Activating mutations in the Kirsten rat sarcoma viral oncogene homolog (KRAS), and epidermal growth factor receptor (EGFR) genes, and the echinoderm microtubule associated protein like 4 – anaplastic lymphoma kinase (EML4–ALK) fusion protein act as oncogenic drivers responsible for tumor growth in a substantial proportion of NSCLC cases [34]. Downstream signal pathways activated in oncogenic-driven lung cancer cells are intricated also in the regulation of PD-L1 expression [35].

About one third of pulmonary adenocarcinoma (ADC) cases harbor activating mutations in KRAS [36]. In pulmonary ADC cell lines, activation of KRAS led to up-regulation of PD-L1 through the stimulation of the mitogen-activated protein kinase (MAPK), causing inhibition of tristetraproline, mediated by p38-dependent phosphorylation [37,38]. Furthermore, downstream signaling by activated RAS caused stabilization of PD-L1 mRNA transcripts [37,38]. Both, tristetraproline inhibition as well as stabilization of PD-L1 transcriptional products eventually led to an increase of PD-L1 expression in pulmonary ADC cell lines [37,38]. Poorly differentiated, grade 3 pulmonary ADC harbor KRAS mutations more often [39] and tend to have a higher level of PD-L1 expression when compared to lower grade ADCs [40]. These tumors also have a higher tumor mutational burden (TMB) reported in studies and a more prominent T-cell infiltrate, which underlines the feasibility of immunotherapeutic approaches in this NSCLC group [41].

Genetic alterations in the EGFR are present in about 10 to 25 % of pulmonary ADC patients, and higher in persons of Asian descent, in woman, and in non-smokers [42–44]. A positive result of PD-L1 immunohistochemistry at baseline assessment has been linked to the presence of EGFR mutations in tumors from lung cancer patients [45]. Activating mutations of EGFR led to up-regulation of PD-L1 expression in tumor cells of NSCLC through activation of several signaling pathways including the JAK/STAT and the phosphatidylinositol 3-kinase / Protein kinase B / mammalian target of rapamycin (PI3K / Akt / mTOR) axis, as well as downstream signaling mediated by activation of MAPK [46–49]. Despite the intricated and interconnected signaling pathways leading to up-regulation of PD-L1 in EGFR-mutated pulmonary ADCs, the level of PD-L1 positivity in these tumors tend to be in the category of low expression [50]. This can be attributed to the relative paucity of TIL the stroma and tumor tissue with the associated lack of external stimuli of PD-L1 expression through INF- g elaboration [50]. Furthermore, ADCs harboring EGFR mutations tend to be genetically less complex when compared to KRAS-mutated NSCLCs, displaying a lower number of non-synonymous mutations per coding area of their genome, and thus having a low TMB [51,52]. Overall, the reported clinical outcomes and response rate to ICIs are poor in patients having EGFR-mutated NSCLCs [50].

High levels of PD-L1 immunoreactivity have been reported in ADCs harboring the EML4–ALK fusion protein [53]. Up-regulation of PD-L1 in NSCLCs with ALK rearrangement is mediated through activation of the PI3K / Akt / mTOR signaling pathway, the MEK / ERK axis, and STAT3 [53–55]. The latter increased the transcription of PD-L1-related genes by directly binding and interacting with the promoter region of the CD274 locus [55].

Amplification of CD274  and PD-L1 expression

Besides the described correlation of PD-L1 expression with the presence of oncogenic driver mutations in tumors, higher levels of PD-L1 positivity were encountered in many cancer types having amplifications of the locus 9p24.1, where CD274 resides [56–58]. Copy number alteration in genes related to PD-L1 have been described also in tumor cells from patients having advanced NSCLC [57]. Inoue et al (2016) identified amplification or polysomy of PD-L1 in 3.1 % and 13.2 % of NSCLCs, respectively [57]. The status of copy number increase of PD-L1 positively correlated with elevation of the number of genes related to the JAK2, as well as with amplification of PD-L2 [57]. Furthermore, multivariant analyses identified polysomy and amplification of PD-L1 as independent factors associated with high level of PD-L1 expression, with a pronounced infiltration by lymphocytes, and with the presence of EGFR mutations in tumors from NSCLC patients [57].

These findings indicate that amplification of PD-L1 is an important mechanism by which tumor cells are able escape immunosurveillance. Therefore, gene copy number evaluation represents a feasible alternative marker, predicting clinical outcome and efficacy of ICI [57].

Regulation of PD-L1 and cancer mesenchymalization

Mesenchymalization is an important concept in modern oncological research, encompassing all molecular programs and cellular mechanisms which cause epithelial cells to lose their well-differentiated phenotype and acquire mesenchymal characteristics [59]. Cancer mesenchymalization develops during the progression of a locally advanced to fully metastatic disease and it has been also implicated in the development of drug resistance to various treatment modalities [60,61]. The multitude of molecular mechanisms responsible for the epithelial to mesenchymal transition (EMT) in cancer cells are involved also in the regulation of PD-L1 [62]. David et al [63] came to conclusion that activation of the transforming growth factor beta 1   (TGF-b1) axis stimulates the transcription of genes related to PD-L1 in  a manner dependent on Smad2. According to their published data, the level of PD-L1 expression positively correlated with the amount of phosphorylated Smad2 present in tumor cells of NSCLC [63]. Treatment of lung cancer cells in vitro or in vivo with the clinical-stage bifunctional agent M7824, which targets PD-L1 as well as TGF-b1, hampered features of mesenchymalization mediated by TGF-b1, including the pos­itivity of mesenchymal markers, proliferation of tumor cells, and resistance to chemotherapy [63].

In another study, Evanno et al [64] observed that TGF-b1-induced EMT in lung cancer cells resulted in an increase of the level of PD-L1 expression, which was mediated by modifications of the histone methylation process. Demethylation of the promoter region related to the PD-L1 locus led to up-regulation of PD-L1 and correlated with down-regulation of epithelial markers such as E-cadherin [64]. Authors of this study suggest that adding epigenetic modifiers to conventional chemotherapy or ICI could improve clinical outcomes and reduce the risk of metastasis [64].

Cumulative published data suggest that cancers which had undergone the most pronounced degree of mesenchymalization demonstrate the highest level of PD-L1 expression: Pleomorphic carcinomas of the lung are reported to have overall PD-L1 positivity within the range of 52 to 90 % [65–67]. Besides cancers with overt histological signs of mesenchymalization, poorly differentiated, grade 3 pulmonary ADC share a high level of PD-L1 expression, underscoring that escape from immune surveillance is a very important early step in the tumor progression process [68].

These results demonstrate the importance of immune escape in cancers undergoing mesenchymalization and point out to the potential therapeutic value and the need of deeper understanding of the molecular mechanisms related to EMT, which are involved also in the regulation of PD-L1 [63].

Desmoplasia and  PD-L1 expression

The activation of the TGF-b1 pathway in tumor associated fibroblasts is also linked to the elaboration of dense collagen fibers and other connective tissue components eventually leading to desmoplastic stroma production [69]. Mariathasan et al [70] demonstrated a significant inverse correlation between the degree of desmoplasia and quantitatively measured amount of TIL within the tumor. They have shown that within the TME, a dense desmoplastic stroma represents a mechanical barrier which excludes immune cells and hinders them from infiltrating the tumor tissue [70]. Tumors flooded with lymphocytes are associated with better prognosis [71–73]. The presence of T-cells in the TME is also necessary to modulate immune characteristics of tumor cells, like PD-L1 expres­sion. Preclinical studies are ongoing, which are testing combinational therapy aiming to block the PD-1 axis as well as reverting TGF-b1-mediated tumor fibrotization, and they yield promising results so far [70].

Expression of PD-L1 under hypoxic conditions

The presence of coagulative necrosis in tumors can be viewed as a morphological marker of severe hypoxic conditions within the TME [66,74], and hypoxic signaling pathways have been shown to be interconnected with various immunological aspects [75]. The presence of necrotic areas in tumors from patients having lung ADC was associated with PD-L1 positivity in tumor cells, and higher PD-1 expression in immune cells [76]. Chung et al [66] have found a similar association in pulmonary pleomorphic carcinomas, where extensive necrosis correlated significantly with high PD-L1 expression in tumor cells (P < 0.001). Furthermore, PD-L1 positivity was highest in cells in the direct vicinity to necrotic areas [66]. Barsoum et al (2014) provided a general mechanistic explanation of this association on the molecular level, by showing that up-regulation of PD-L1 in cancer cells, which were exposed to a hypoxic environment, was mediated by activation of the transcriptional factor hypoxia-inducible factor-1a (HIF1A) [75]. Noonan et al [77] demonstrated a significant, rapid, and selective increase in PD-L1 expression in cancer cells cultivated under hypoxic conditions for 24 hours. In the same study, they showed direct binding of HIF1A to the transcriptionally active hypoxia-responsible elements of the promoter region of the PD-L1 [77]. The positive correlation between the expression of PD-L1 and HIF1A was corroborated in studies of tumors from lung cancer patients having advanced ADC [78], or pleomorphic carcinomas [66]. Other hypoxic signaling pathways are also implicated in the regulation of PD-L1 in pulmonary ADC, as was shown by Koh et al [78]: They have found that PD-L1 significantly correlated with HIF1A, carbonic anhydrase IX, vascular endothelial growth factor A (VEGFA), and glucose transporter 1 (GLUT1) on the protein as well as on the mRNA level [78]. Patients with low level of PD-L1 expression combined with low GLUT1 expression in tumors displayed longer overall survival, which suggest the additional prognostic value of these two markers [78]. The proangiogenic factor VEGFA has also additional effects on the activity of immune responses directed against tumor growth, since it hampers antigen presentation, stimulates infiltration of the TME with regulatory T-cells, and causes up-regulation of PD-L1 in TILs [79]. Regarding driver mutations, immunohistochemical analysis in studies showed a positive correlation between the expression of NF-kB, HIF1A, and PD-L1 in NSCLCs having mutations of the EGFR [80]. Increase of PD-L1 expression was also mediated by up-regulation of HIF1A and/or STAT3 in lung ADC patients harboring EML4-ALK translocations [81].

These findings underscore the importance of hypoxic signaling pathways in the regulation of PD-L1 expression, and concomitant blockade of hypoxic signaling pathways as well as the PD-1/PD-L1 axis represents, therefore, a perspective combined immunotherapeutic approach.

 

Conclusions

Evaluation of PD-L1 by means of immunohistochemistry is still the mainstay marker used to predict clinical efficacy of ICI in patient with advanced NSCLC. PD-L1 negativity does not always preclude effectiveness and survival benefit of immunotherapy, and some highly PD-L1 positive NSCLC have been reported as being irresponsive to ICI. These inconsistent results possibly stem from the interaction of the PD-1 signaling pathway with numerous factors within the TME, which were, probably, not taken comprehensively into account in clinical studies. A deeper understanding of the mechanisms which regulate the expression of PD-L in the TME is crucial to create therapeutic algorithms and improve strategies which harness the immune system to fight cancer.

Dedication

This publication was created thanks to the support of the Operational Program Integrated Infrastructure for the project Integrative strategy in development of personalized medicine of selected malignant tumors and its impact on quality of life, IMTS: 313011V446, co-financed by the European Regional Development Fund.

The authors declare that they have no potential conflicts of interest concerning drugs, products, or services used in the study.
Autoři deklarují, že v souvislosti s předmětem studie nemají žádné komerční zájmy.
The Editorial Board declares that the manuscript met the ICMJE recommendation for biomedical papers.
Redakční rada potvrzuje, že rukopis práce splnil ICMJE kritéria pro publikace zasílané do biomedicínských časopisů.

MUDr. Alžbeta Blichárová, PhD
Ústav patológie UPJŠ LF a UNLP
Rastislavova 43
040 01 Košice
Slovenská Republika

e-mail: alzbeta.blicharova@upjs.sk

Submitted/Obdŕžané: 4. 5. 2021
Accepted/Prijaté: 12. 7. 2021


Zdroje

1. Mallett G, Laurence A, Amarnath S. Programmed cell death-1 receptor (PD-1) -mediated regulation of innate lymphoid cells. Int J Mol Sci 2019; 20 (11): 2836. doi: 10.3390/ijms20112836.

2. Zatloukalová P, Pjechová M, Babčanová S et al. The role of PD-1/PD-L1 signaling pathway in antitumor immune response. Klin Onkol 2016; 29 (Suppl 4): 4S72–4S77. doi: 10.14735/amko20164S72.

3. Zou W, Wolchok JD, Chen L. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: mechanisms, response biomarkers, and combinations. Sci Transl Med 2016; 8 (328): 328rv4. doi: 10.1126/scitranslmed.aad7118.

4. Koubková L. Immunotherapy of bronchogenic carcinoma and its perspectives. Klin Onkol 2015; 28 (Suppl 4): 4S77–4S81. doi: 10.14735/amko20154S77.

5. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012; 12 (4): 252–264. doi: 10.1038/nrc3239.

6. Gridelli C, Ardizzoni A, Barberis et al. Predictive biomarkers of immunotherapy for non-small cell lung cancer: results from an experts panel meeting of the Italian association of thoracic oncology. Transl Lung Cancer Res 2017; 6 (3): 373–386. doi: 10.21037/tlcr.2017.05.09.

7. Garon EB, Rizvi NA, Hui et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med 2015; 372 (21): 2018–2028. doi: 10.1056/NEJMoa1501824.

8. Fehrenbacher L, Spira A, Ballinger M 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 (10030): 1837–1846. doi: 10.1016/S0140-6736 (16) 00587-0.

9. Borghaei H, Paz-Ares L, Horn L et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med 2015; 373 (17): 1627–1639. doi: 10.1056/NEJMoa1507643.

10. Herbst RS, Baas P, Kim D-W et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet 2016; 387 (10027): 1540–1550. doi: 10.1016/S0140-6736 (15) 01281-7.

11. Hui R, Garon EB, Goldman et al. Pembrolizumab as first-line therapy for patients with PD-L1-positive advanced non-small cell lung cancer: a phase 1 trial. Ann Oncology 2017; 28 (4): 874–881. doi: 10.1093/annonc/mdx008.

12. Wang C, Wang H, Wang L. Biomarkers for predicting the efficacy of immune checkpoint inhibitors. J Cancer 2022; 13 (2): 481–495. doi: 10.7150/jca.65012.

13. Lagos GG, Izar B, Rizvi NA. Beyond tumor PD-L1: emerging genomic biomarkers for checkpoint inhibitor immunotherapy. Am Soc Clin Oncol Educ Book 2020; 40: 1–11. doi: 10.1200/EDBK_289967.

14. Chen J, Jiang CC, Jin L et al. Regulation of PD-L1: a novel role of pro-survival signalling in cancer. Ann Oncol 2016; 27 (3): 409–416. doi: 10.1093/annonc/mdv615.

15. Jiang Y, Zhan H. Communication between EMT and PD-L1 signaling: new insights into tumor immune evasion. Cancer Lett 2020; 468: 72–81. doi: 10.1016/j.canlet.2019.10.013. 2X.2015.1108514.

16. Sun C, Mezzadra R, Schumacher TN et al. Regulation and Function of the PD-L1 Checkpoint. Immunity 2018; 48 (3): 434–452. doi: 10.1016/j.immuni.2018.03. 014.

17. Jiang Y, Chen M, Nie H et al. PD-1 and PD-L1 in cancer immunotherapy: clinical implications and future considerations. Hum Vaccin Immunother 2019; 15 (5):  1111–1122. doi: 10.1080/21645515.2019.1571892.

18. Lamberti G, Sisi M, Andrini E et al. The mechanisms of PD-L1 regulation in non-small-cell lung cancer (NSCLC): which are the involved players? Cancers 2020; 12 (11): 3129. doi: 10.3390/cancers12113129.

19. Garcia-Diaz A, Shin DS, Moreno BH et al. Interferon receptor signaling pathways regulating PD-L1 and PD-L2 expression. Cell Rep 2017; 19 (6): 1189–1201. doi: 10.1016/j.celrep.2017.04.031.

20. Wang X, Yang L, Huang F et al. Inflammatory cytokines IL-17 and TNF-a up-regulate PD-L1 expression in human prostate and colon cancer cells. Immunol Lett 2017; 184: 7–14. doi: 10.1016/j.imlet.2017.02.006.

21. Zhang W, Liu Y, Yan Z et al. IL-6 promotes PD-L1 expression in monocytes and macrophages by decreasing protein tyrosine phosphatase receptor type O expression in human hepatocellular carcinoma. J Immunother Cancer 2020; 8 (1): e000285. doi: 10.1136/jitc-2019-000285.

22. Shen MJ, Xu LJ, Yang L et al. Radiation alters PD-L1/NKG2D ligand levels in lung cancer cells and leads to immune escape from NK cell cytotoxicity via IL-6-MEK/Erk signaling pathway. Oncotarget 2017; 8 (46): 80506–80520. doi: 10.18632/oncotarget.19193.

23. Jiang C, Yuan F, Wang J et al. Oral squamous cell carcinoma suppressed antitumor immunity through induction of PD-L1 expression on tumor-associated macrophages. Immunobio­logy 2017; 222 (4): 651–657. doi: 10.1016/j.imbio­.2016.12.002.

24. Carbotti G, Barisione G, Airoldi I et al. IL-27 induces the expression of IDO and PD-L1 in human cancer cells. Oncotarget 2015; 6 (41): 43267–43280. doi: 10.18632/oncotarget.6530.

25. Yi M, Niu M, Xu L et al. Regulation of PD-L1 expression in the tumor microenvironment. J Hematol Oncol 2021; 14 (1): 10. doi: 10.1186/s13045-020-01027-5.

26. Hendry S, Salgado R, Gevaert T et al. Assessing tumor-infiltrating lymphocytes in solid tumors: a practical review for pathologists and proposal for a standardized method from the International Immuno-Oncology Biomarkers Working Group: Part 2: TILs in melanoma, gastrointestinal tract carcinomas, non-small cell lung carcinoma and mesothelioma, endometrial and ovarian carcinomas, squamous cell carcinoma of the head and neck, genitourinary carcinomas, and primary brain tumors. Adv Anat Pathol 2017; 24 (6): 311–335. doi: 10.1097/PAP.0000000000000161.

27. Salgado R, Denkert C, Demaria S et al. The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014. Ann Oncol 2015; 26 (2): 259–271. doi: 10.1093/annonc/mdu450.

28. Wong PF, Wei W, Smithy JW et al. Multiplex quantitative analysis of tumor-infiltrating lymphocytes and immunotherapy outcome in metastatic melanoma. Clin Cancer Res 2019; 25 (8): 2442–2449. doi: 10.1158/1078-0432.CCR-18-2652.

29. Paijens ST, Vledder A, de Bruyn M et al. Tumor-infiltrating lymphocytes in the immunotherapy era. Cell Mol Immunol 2021; 18 (4): 842–859. doi: 10.1038/s41423-020-00565-9.

30. Karachaliou N, Gonzalez-Cao M, Crespo G 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: 175883401774974. doi: 10.1177/1758834017749 748.

31. Ayers M, Lunceford J, Nebozhyn M et al. IFN- g–related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest 2017; 127 (8): 2930–2940. doi: 10.1172/ JCI91190.

32. Fumet J-D, Richard C, Ledys 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 (8): 950–960. doi: 10.1038/s41416-018-0220-9.

33. Gettinger SN, Choi J, Mani N et al. A dormant TIL phenotype defines non-small cell lung carcinomas sensitive to immune checkpoint blockers. Nat Commun 2018; 9 (1): 3196. doi: 10.1038/s41467-018-05032-8.

34. Brody R, Zhang Y, Ballas M et al. PD-L1 expression in advanced NSCLC: insights into risk stratification and treatment selection from a systematic literature review. Lung Cancer 2017; 112: 200–215. doi: 10.1016/j.lungcan.2017.08.005.

35. Lingling Z, Jiewei L, Li W et al. Molecular regulatory network of PD-1/PD-L1 in non-small cell lung cancer. Pathol Res Prac 2020; 216 (4): 152852. doi: 10.1016/j.prp.2020.152852.

36. Dogan S, Shen R, Ang DC et al. Molecular epidemiology of EGFR and KRAS mutations in 3,026 lung adenocarcinomas: higher susceptibility of women to smoking-related KRAS –mutant cancers. Clin Cancer Res 2012; 18 (22): 6169–6177. doi: 10.1158/1078-0432.CCR-11-3265.

37. Coelho MA, de Carné Trécesson S, Rana S et al. Oncogenic RAS signaling promotes tumor immunoresistance by stabilizing PD-L1 mRNA. Immunity 2017; 47 (6): 1083–1099.e6. doi: 10.1016/j.immuni.2017.11.016.

38. Chen N, Fang W, Lin Z et al. KRAS mutation-induced upregulation of PD-L1 mediates immune escape in human lung adenocarcinoma. Cancer Immunol Immunother 2017; 66 (9): 1175–1187. doi: 10.1007/s00262-017-2005-z.

39. Rekhtman N, Ang DC, Riely GJ et al. KRAS mutations are associated with solid growth pattern and tumor-infiltrating leukocytes in lung adenocarcinoma. Mod Pathol 2013; 26 (10): 1307–1319. doi: 10.1038/modpathol.2013.74.

40. Forest F, Casteillo F, Da Cruz V et al. Heterogeneity of PD-L1 expression in lung adenocarcinoma metastasis is related to histopathological subtypes. Lung Cancer 2021; 155: 1–9. doi: 10.1016/j.lungcan.2021.02.032.

41. Brody R, Zhang Y, Ballas M et al. PD-L1 expression in advanced NSCLC: insights into risk stratification and treatment selection from a systematic literature review. Lung Cancer 2017; 112: 200–215. doi: 10.1016/j.lungcan.2017.08.005.

42. Sonobe M, Manabe T, Wada H et al. Mutations in the epidermal growth factor receptor gene are linked to smoking-independent, lung adenocarcinoma. Br J Cancer 2005; 93 (3): 355–363. doi: 10.1038/sj.bjc.6602 707.

43. Sharma SV, Bell DW, Settleman J et al. Epidermal growth factor receptor mutations in lung cancer. Nat Rev Cancer 2007; 7 (3): 169–181. doi: 10.1038/nrc2088.

44. Shi Y, Au JS-K, Thongprasert S et al. A prospective, molecular epidemiology study of EGFR mutations in Asian patients with advanced non-small-cell lung cancer of adenocarcinoma histology (PIONEER). J Thorac Oncol 2014; 9 (2): 154–162. doi: 10.1097/JTO.0000000000000 033.

45. Saruwatari K, Ikemura S, Sekihara K et al. Aggressive tumor microenvironment of solid predominant lung adenocarcinoma subtype harboring with epidermal growth factor receptor mutations. Lung Cancer 2016; 91: 7–14. doi: 10.1016/j.lungcan.2015.11.012.

46. Chen N, Fang W, Zhan J et al. Upregulation of PD-L1 by EGFR activation mediates the immune escape in EGFR-driven NSCLC: implication for optional immune targeted therapy for NSCLC patients with EGFR mutation. J Thorac Oncol 2015; 10 (6): 910–923. doi: 10.1097/JTO.00000 00000000500.

47. Akbay EA, Koyama S, Carretero J et al. Activation of the PD-1 pathway contributes to immune escape in EGFR-driven lung tumors. Cancer Discov 2013; 3 (12): 1355–1363. doi: 10.1158/2159-8290.CD-13-0310.

48. Lastwika KJ, Wilson W, Li QK et al. Control of PD-L1 expression by oncogenic activation of the AKT–mTOR pathway in non-small cell lung cancer. Cancer Res 2016; 76 (2): 227–238. doi: 10.1158/0008-5472.CAN-14- 3362.

49. Zhang N, Zeng Y, Du W et al. The EGFR pathway is involved in the regulation of PD-L1 expression via the IL-6/JAK/STAT3 signaling pathway in EGFR-mutated non-small cell lung cancer. Int J Oncol 2016; 49 (4): 1360–1368. doi: 10.3892/ijo.2016.3632.

50. Gainor JF, Shaw AT, Sequist LV et al. EGFR mutations and ALK rearrangements are associated with low response rates to PD-1 pathway blockade in non-small cell lung cancer: a retrospective analysis. Clin Cancer Res 2016; 22 (18): 4585–4593. doi: 10.1158/1078-0432.CCR-15-3101.

51. Meléndez B, Van Campenhout C, Rorive S et al. Methods of measurement for tumor mutational burden in tumor tissue. Transl Lung Cancer Res 2018; 7 (6): 661–667. doi: 10.21037/tlcr.2018.08.02.

52. Toki MI, Mani N, Smithy JW et al. Immune marker profiling and programmed death ligand 1 expression across NSCLC mutations. J Thorac Oncol 2018; 13 (12):  1884–1896. doi: 10.1016/j.jtho.2018.09.012.

53. Ota K, Azuma K, Kawahara A et al. Induction of PD-L1 expression by the EML4–ALK oncoprotein and downstream signaling pathways in non-small cell lung cancer. Clin Cancer Res 2015; 21 (17): 4014–4021. doi: 10.1158/1078-0432.CCR-15-0016.

54. Roussel H, De Guillebon E, Biard L et al. Composite bio­markers defined by multiparametric immunofluorescence analysis identify ALK-positive adenocarcinoma as a potential target for immunotherapy. Oncoimmunology 2017; 6 (4): e1286437. doi: 10.1080/2162402X.2017.1286 437.

55. Koh J, Jang J-Y, Keam B et al. EML4-ALK enhances programmed cell death-ligand 1 expression in pulmonary adenocarcinoma via hypoxia-inducible factor (HIF) -1a and STAT3. Oncoimmunology 2016; 5 (3): e1108514. doi: 10.1080/2162402X.2015.1108514.

56. Ikeda S, Goodman AM, Cohen PR et al. Metastatic basal cell carcinoma with amplification of PD-L1: exceptional response to anti-PD1 therapy. NPJ Genom Med 2016; 1: 16037. doi: 10.1038/npjgenmed.2016.37.

57. Inoue Y, Yoshimura K, Mori K et al. Clinical significance of PD-L1 and PD-L2 copy number gains in non-small-cell lung cancer. Oncotarget 2016; 7 (22): 32113–32128. doi: 10.18632/oncotarget.8528.

58. Budczies J, Bockmayr M, Denkert C et al. Pan-cancer analysis of copy number changes in programmed death-ligand 1 (PD-L1, CD274) – associations with gene expression, mutational load, and survival: pan-cancer analysis of PD-L1 CNAs. Genes Chromosomes Cancer 2016; 55 (8): 626–639. doi: 10.1002/gcc.22365.

59. Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. J Clin Invest 2009; 119 (6): 1420–1428. doi: 10.1172/JCI39104.

60. David JM, Dominguez C, Palena C. Pharmacological and immunological targeting of tumor mesenchymalization. Pharmacol Ther 2017; 170: 212–225. doi: 10.1016/j.pharmthera.2016.11.011.

61. Goswami MT, Reka AK, Kurapati H et al. Regulation of complement-dependent cytotoxicity by TGF-b-induced epithelial-mesenchymal transition. Oncogene 2016; 35 (15): 1888–1898. doi: 10.1038/onc.2015.258.

62. Chen L, Gibbons DL, Goswami S et al. Metastasis is regulated via microRNA-200/ZEB1 axis control of tumour cell PD-L1 expression and intratumoral immunosuppression. Nat Commun 2014; 5: 5241. doi: 10.1038/ncomms 6241.

63. David JM, Dominguez C, McCampbell KK et al. A novel bifunctional anti-PD-L1/TGF-b Trap fusion protein (M7824) efficiently reverts mesenchymalization of human lung cancer cells. Oncoimmunology 2017; 6 (10): e1349589. doi: 10.1080/2162402X.2017.1349 589.

64. Evanno E, Godet J, Piccirilli N et al. Tri-methylation of H3K79 is decreased in TGF-b1-induced epithelial-to-mesenchymal transition in lung cancer. Clin Epigenet 2017; 9: 80. doi: 10.1186/s13148-017-0380-0.

65. Yvorel V, Patoir A, Casteillo F et al. PD-L1 expression in pleomorphic, spindle cell and giant cell carcinoma of the lung is related to TTF-1, p40 expression and might indicate a worse prognosis. PLoS One 2017; 12 (10): e0180346. doi: 10.1371/journal.pone.0180346.

66. Chang Y-L, Yang C-Y, Lin M-W et al. High co-expression of PD-L1 and HIF-1a correlates with tumour necrosis in pulmonary pleomorphic carcinoma. Eur J Cancer 2016; 60: 125–135. doi: 10.1016/j.ejca.2016.03. 012.

67. Vieira T, Antoine M, Hamard C et al. Sarcomatoid lung carcinomas show high levels of programmed death ligand-1 (PD-L1) and strong immune-cell infiltration by TCD3 cells and macrophages. Lung Cancer 2016; 98:  51–58. doi: 10.1016/j.lungcan.2016.05.013.

68. Ng Kee Kwong F, Laggner U, McKinney O et al. Expression of PD-L1 correlates with pleomorphic morphology and histological patterns of non-small-cell lung carcinomas. Histopathology 2018; 72 (6): 1024–1032. doi: 10.1111/his.13466.

69. Tancoš V, Grendár M, Farkašová A et al. Programmed death-ligand 1 expression in non-small cell lung carcinoma bio­psies and its association with tumor infiltrating lymphocytes and the degree of desmoplasia. Klin Onkol 2020; 33 (1): 55–65. doi: 10.14735/amko202 055.

70. Mariathasan S, Turley SJ, Nickles D et al. TGFb attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018; 554 (7693): 544–548. doi: 10.1038/nature25501.

71. Schalper KA, Brown J, Carvajal-Hausdorf D et al. Objective measurement and clinical significance of TILs in non-small cell lung cancer. J Natl Cancer Inst 2015; 107 (3): dju435. doi: 10.1093/jnci/dju435.

72. Stanton SE, Disis ML. Clinical significance of tumor-infiltrating lymphocytes in breast cancer. J Immunother Cancer 2016; 4: 59. doi: 10.1186/s40425-016-01 65-6.

73. Maibach F, Sadozai H, Seyed Jafari SM et al. Tumor-infiltrating lymphocytes and their prognostic value in cutaneous melanoma. Front Immunol 2020; 11: 2105. doi: 10.3389/fimmu.2020.02105.

74. Zhou G, Dada LA, Wu M et al. Hypoxia-induced alveolar epithelial-mesenchymal transition requires mitochondrial ROS and hypoxia-inducible factor 1. Am J Physiol Lung Cell Mol Physiol 2009; 297 (6): L1120–L1130. doi: 10.1152/ajplung.00007.2009.

75. Barsoum IB, Smallwood CA, Siemens DR et al. A mechanism of hypoxia-mediated escape from adaptive immunity in cancer cells. Cancer Res 2014; 74 (3): 665–674. doi: 10.1158/0008-5472.CAN-13-0992.

76. Reiniger L, Téglási V, Pipek O et al. Tumor necrosis correlates with PD-L1 and PD-1 expression in lung adenocarcinoma. Acta Oncol 2019; 58 (8): 1087–1094. doi: 10.1080/0284186X.2019.1598575.

77. Noman MZ, Desantis G, Janji B et al. PD-L1 is a novel direct target of HIF-1a, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J Exp Med 2014; 211 (5): 781–790. doi: 10.1084/jem.20131916.

78. Koh YW, Lee SJ, Han J-H et al. PD-L1 protein expression in non-small-cell lung cancer and its relationship with the hypoxia-related signaling pathways: a study based on immunohistochemistry and RNA sequencing data. Lung Cancer 2019; 129: 41–47. doi: 10.1016/j.lungcan.2019.01.004.

79. Voron T, Colussi O, Marcheteau E et al. VEGF-A modulates expression of inhibitory checkpoints on CD8+ T cells in tumors. J Exp Med 2015; 212 (2): 139–148. doi: 10.1084/jem.20140559.

80. Guo R, Li Y, Wang Z et al. Hypoxia-inducible factor-1a and nuclear factor-kB play important roles in regulating programmed cell death ligand 1 expression by epidermal growth factor receptor mutants in non-small-cell lung cancer cells. Cancer Sci 2019; 110 (5): 1665–1675. doi: 10.1111/cas.13989.

81. Koh J, Jang J-Y, Keam B et al. EML4-ALK enhances programmed cell death-ligand 1 expression in pulmonary adenocarcinoma via hypoxia-inducible factor (HIF) -1a and STAT3. Oncoimmunology 2016; 5 (3): e1108514. doi: 10.1080/2162402X.2015.1108514.

Štítky
Detská onkológia Chirurgia všeobecná Onkológia

Článok vyšiel v časopise

Klinická onkologie

Číslo 5

2022 Číslo 5
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Eozinofilní granulomatóza s polyangiitidou
nový kurz
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#