Abstract
According to the latest Global Cancer Observatory (GLOBOCAN) 2022 estimates, lung cancer remains the second most commonly diagnosed cancer worldwide, with approximately 2.48 million new cases (11.4% of all cancer diagnoses) and over 1.86 million deaths (18.2% of all cancer-related deaths), making it the leading cause of cancer mortality globally. It has been reported that programmed death ligand 1 (PD-L1) is confirmed to interact with the tumor microenvironment (TME) to mediate the immune escape of lung cancer. PD-L1, which is highly expressed in lung cancer cells, activates the programmed death receptor 1 (PD-1)/PD-L1 signaling pathway by binding to PD-1, thereby inhibiting the function of lymphocytes and the release of cytokines, inducing activated lymphocyte apoptosis, resisting the killing effect of lymphocytes, ultimately leading to immune escape in lung cancer. PD-L1 inhibitors, a hot spot in tumor immunotherapy, can restore the activity of T cells, thereby enhancing the body’s immune response, and ultimately enabling the immune system to effectively recognize and kill lung cancer cells, thereby enabling lung cancer patients to achieve long-term tumor remission. At present, a variety of PD-L1 inhibitors have been approved for application and have achieved good clinical efficacy in the treatment of lung cancer. This article reviews the research progress of the interaction between PD-L1 and the TME to mediate immune escape from lung cancer and the role of PD-L1 inhibitors in the treatment of lung cancer.
Keywords: Lung cancers, immune responses, immune evasion, mechanism, research progress
Introduction
Lung cancer is one of the common malignant tumors worldwide, with high metastasis rate and recurrence rate. Despite advances in treatment, the 5-year survival rate for lung cancer remains below 20%, largely due to late-stage diagnosis and high metastatic potential (1). Although targeted therapies offer specificity and fewer side effects, their efficacy can be compromised by gene mutations and drug resistance. Immunotherapy, particularly monoclonal antibodies such as programmed death receptor-1 (PD-1)/programmed death ligand 1 (PD-L1) inhibitors, represents a distinct treatment modality. While the initial cost of these agents was high, recent years have seen a reduction in price due to the availability of biosimilars and commercial agreements, improving accessibility for broader patient populations (2). With the development of immunology and molecular biology, immunotherapy has become a research hotspot. PD-1, first identified by Ishida et al. in 1992, and its ligand PD-L1 are negative immunomodulatory molecules that have attracted increasing attention in recent years due to their crucial role in the immune escape mechanism of tumor cells (3). The activation of PD-1/PD-L1 signaling can lead to the formation of immunosuppressive tumor microenvironment (TME), which enables tumor cells to evade the monitoring and killing of the immune system (4).
At present, the core of tumor immunotherapy is to activate the anti-tumor effect of T lymphocytes and improve the killing ability of tumor cells. A recent study has found that PD-L1 is highly expressed in a variety of tumors and inhibits the proliferation and activation of lymphocytes and anti-tumor immunity by binding to PD-1 molecules on the surface of tumor-infiltrating lymphocytes (TILs), which is one of the important reasons for tumor immune escape (5). Therefore, it is of great clinical significance to study the mechanism of PD-1/PD-L1 signaling in tumor immune escape and provide reliable evidence and ideas for tumor immunotherapy. This review summarizes and analyzes the mechanism and research progress of PD-1/PD-L1 in tumor immune escape. This article is presented as a narrative review, aiming to integrate key clinical trial results, molecular mechanisms, and evolving therapeutic strategies related to PD-1/PD-L1 inhibitors in lung cancer. A narrative format was chosen to provide both a broad perspective and an in-depth interpretation of rapidly developing areas that may not yet be captured in systematic analyses.
Molecular mechanism of PD-1/PD-L1 on tumor immune escape of lung cancer
PD-1 is a transmembrane receptor on T cells, which is named programmed death 1 receptor because it is involved in the process of cell apoptosis (3). PD-1 is a transmembrane glycoprotein of approximately 55 KDa encoded by the PDCD1 gene located on chromosome 2Q37.35. PD-1 is mainly expressed on the membrane surface of T cells, B cells, natural killer (NK) cells, and a variety of tumor cells (6). It has been confirmed that the ligands of PD-1 are composed of PD-L1 and PD-L2, both of which belong to the B7 superfamily, and the coding genes are located on human chromosome 9P24.2. PD-L1 (also known as B7-H1, CD274) is the main ligand of PD-1, which is widely expressed on the surface of antigen presenting cells (APCs), macrophages, activated T cells and B cells, monocytes, and endothelial cells (7). However, the expression of PD-L2 (also known as B7-DC and CD273) was limited, mainly expressed in macrophages, dendritic cells (DCs), and mast cells.
PD-1 binds to PD-L1 and induces tyrosine phosphorylation of the immunoreceptor tyrosine-based switch motif (ITSM) in the intracellular region of PD-1 mainly through T cell receptor (TCR) signaling, which then recruits protein tyrosine phosphatase 1 (SHP-1) and protein tyrosine phosphatase 2 (SHP-2) in the Src homeodomain (8). SHP-2 dephosphorylates TCR-associated CD3ζ and Zeta chain-associated protein kinases (ZAP70), thereby inhibiting T-cell activation signals, including phosphoinositide 3-kinase (PI3K) and its downstream protein kinase Akt, glucose metabolism disorders, and interleukin-2 secretion inhibition (9). The negative signal mediated by this process can effectively block the proliferation and activation of T lymphocytes and the secretion of cytokines, and negatively regulate the immune response (10). The activated T cells show a state of “hyper exhaustion”, which reduces the activity of effector T cells and terminates the immune response. Under normal circumstances, excessive inflammatory reaction or autoimmune reaction can be prevented (11). The significant expression of PD-L1 on the surface of lung cancer cells and its binding to PD-1 on the surface of lymphocytes will weaken the body’s anti-tumor immune function, form an immune microenvironment conducive to tumor survival, promote tumor immune escape, and participate in the occurrence and development of tumors (12,13) (Figure 1). Recent studies have shown that both PD-1/PD-L1 can participate in tumor immune escape through a variety of molecular signaling pathways (14-17).
Figure 1.
Mechanism of action of PD-1/PD-L1 inhibitors. APC, antigen-presenting cells; MHC, major histocompatibility complex; PD-1, programmed death receptor 1; PD-L1, programmed death ligand 1; TCR, T cell receptor.
Hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF) signaling pathways
Hypoxia is a common feature of the TME in lung cancer, contributing significantly to immune escape and therapeutic resistance. Under hypoxic conditions, the transcription factor HIF-1α is stabilized and translocated into the nucleus, where it binds to hypoxia response elements (HREs) in the promoter region of several target genes, including PD-L1 (18). This leads to the upregulation of PD-L1 expression in tumor cells, thereby enhancing their ability to evade cytotoxic T lymphocyte (CTL) surveillance by engaging the PD-1 receptor and inducing CTL apoptosis or exhaustion (19).
In addition to PD-L1, HIF-1α also promotes the transcription of VEGF, a key driver of tumor angiogenesis (18). VEGF not only facilitates neovascularization but also plays a pivotal role in shaping an immunosuppressive microenvironment. Elevated VEGF levels inhibit DC maturation, impair antigen presentation, promote the recruitment of regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), and reduce the infiltration and activation of effector T cells in tumor tissue (19,20). These effects collectively result in an immunologically “cold” TME, which is typically less responsive to immune checkpoint inhibitors such as anti-PD-1/PD-L1 antibodies.
Moreover, clinical studies have suggested that patients with tumors exhibiting high HIF-1α or VEGF expression may display primary resistance to PD-1/PD-L1 blockade. To address this, combination strategies involving anti-VEGF agents (e.g., bevacizumab) and PD-1/PD-L1 inhibitors (e.g., atezolizumab) have been developed and shown to improve therapeutic outcomes by simultaneously normalizing the vasculature and restoring T cell infiltration (18,20). Thus, the HIF-1α-VEGF axis plays a central role in mediating tumor immune escape and influencing the sensitivity or resistance to PD-1/PD-L1-targeted immunotherapy (Figure 2).
Figure 2.
Hypoxia-induced regulation of PD-L1 expression and immune evasion via the HIF-1α/VEGF pathway. CTL, cytotoxic T lymphocyte; DC, dendritic cell; HIF-1α, hypoxia inducible factor-1α; MDSC, myeloid-derived suppressor cell; PD-1, programmed death receptor 1; PD-L1, programmed death ligand 1; Treg, regulatory T cell; VEGF, vascular endothelial growth factor.
Tumor-derived transforming growth factor-β (TGF-β) signaling pathway
TGF-β is a multifunctional cytokine with potent immunosuppressive properties, playing a key role in promoting immune tolerance and tumor immune escape. Within the TME, TGF-β is secreted by tumor-associated DCs, cancer cells, and Tregs, and contributes to immunosuppression through several mechanisms (20).
One key pathway involves the suppression of effector T cell function via transcriptional reprogramming. TGF-β inhibits the expression of special AT-rich binding protein 1 (SATB1), a chromatin organizer that normally acts to repress PD-1 transcription in activated T cells (21). Specifically, TGF-β activates SMAD signaling cascades that bind to the SATB1 promoter and inhibit its expression, thereby preventing SATB1 from recruiting the NuRD histone deacetylase complex to PD-1 enhancer regions (22). This epigenetic derepression leads to upregulated PD-1 expression, promoting T cell exhaustion and loss of cytotoxic function in the TME.
In addition, TGF-β exerts physical barriers to immune infiltration by inducing stromal remodeling and extracellular matrix deposition. This leads to the formation of the so-called “immune-excluded phenotype”, in which CD8+ T cells are trapped in the stromal periphery and fail to infiltrate the tumor core. Such tumors typically exhibit poor responses to PD-1/PD-L1 inhibitors, not due to lack of antigenicity, but due to spatial segregation of immune effectors from target cells.
To overcome this form of resistance, a recent preclinical and clinical study has evaluated dual blockade of TGF-β and PD-L1, including fusion proteins such as SHR-1701, which simultaneously targets the PD-L1 and TGF-β pathways (20). These strategies have shown promise in reversing T cell exclusion and enhancing the efficacy of immune checkpoint blockade. Overall, TGF-β-driven immune exclusion and PD-1 upregulation represent major mechanisms of adaptive immune resistance, and targeting this axis may significantly improve patient response to immunotherapy (Figure 3).
Figure 3.

TGF-β signaling promotes PD-1 expression and immune exclusion. GSK-3β, glycogen synthase kinase 3 beta; PD-1, programmed death receptor 1; PD-L1, programmed death ligand 1; TGF-β, transforming growth factor-β.
Glycogen synthase kinase-3 (GSK-3) signaling pathway
GSK-3 is a constitutively active serine/threonine kinase involved in diverse cellular functions, including metabolism, cell cycle regulation, and immune modulation. In the context of T cell immunity, GSK-3 plays a critical role in regulating PD-1 expression and T cell exhaustion. Upon TCR activation, GSK-3 is transiently inhibited, which permits the transcriptional upregulation of T-bet (T-box transcription factor TBX21), a key transcriptional repressor of PD-1 (PDCD1) (23,24).
T-bet directly binds to the promoter region of PDCD1 and suppresses its transcription, thus maintaining effector T cell function and preventing exhaustion. However, in TILs, chronic antigen stimulation and aberrant TME signals can restore GSK-3 activity, which suppresses T-bet levels and thereby sustains PD-1 expression. This shift leads to a progressive loss of T cell cytotoxicity, cytokine secretion, and proliferation capacity—a hallmark of exhausted T cells (24).
Experimental inhibition of GSK-3 using small molecule inhibitors or siRNAs has been shown to restore T cell function by reducing PD-1 expression and enhancing interferon-gamma (IFN-γ) and granzyme B production in CD8+ CTLs. Importantly, Taylor et al. confirmed that GSK-3 inactivation increases the binding affinity of T-bet to the PDCD1 promoter, further validating the regulatory axis (24). In in vivo tumor models, pharmacological GSK-3 inhibition synergized with PD-1 blockade to improve antitumor efficacy, suggesting that GSK-3 inhibition can act as a sensitizer to immune checkpoint therapy (25).
Thus, GSK-3 represents a critical upstream regulator of PD-1 expression and T cell exhaustion, and targeting this pathway offers a promising strategy to reverse resistance and enhance the clinical efficacy of PD-1/PD-L1 inhibitors (Figure 4).
Figure 4.

GSK-3-T-bet-PD-1 axis regulates T cell exhaustion and checkpoint sensitivity. CTL, cytotoxic T lymphocyte; GSK-3, glycogen synthase kinase-3; HIF-1α, hypoxia inducible factor-1α; MDSC, myeloid-derived suppressor cell; PD-1, programmed death receptor 1; PD-L1, programmed death ligand 1; Treg, regulatory T cell; VEGF, vascular endothelial growth factor.
Application of PD-1/PD-L1 inhibitors in lung cancer
Table 1 shows the clinical treatment and indications of PD-1/PD-L1 inhibitors widely used in advanced lung cancer in China and other countries.
Table 1. The clinical treatment and indications of PD-1/PD-L1 inhibitors used in advanced lung cancer.
| Drug name | Target | R&D institutions | Antibody type | Pivotal trial | Global clinical trials | Chinese clinical trials | Approved | Other major approved indications |
|---|---|---|---|---|---|---|---|---|
| Nivolumab | PD-1 | Ono Pharmaceutical | IgG4 | CheckMate-017, CheckMate-057 | 75 | 7 | US [2014], China [2018] | Metastatic melanoma, adjuvant melanoma therapy, metastatic non-small cell lung cancer, classical Hodgkin lymphoma, renal cell carcinoma, head and neck squamous cell carcinoma, hepatocellular carcinoma, urethral carcinoma, bladder cancer, mismatch repair deficiency colon cancer |
| Pembrolizumab | PD-1 | Merck | IgG4 | KEYNOTE-024, KEYNOTE-042, KEYNOTE-010 |
103 | 15 | US [2017], China [2018] | High microsatellite unstable tumors, melanoma, non-small cell lung cancer, classic Hodgkin lymphoma, head and neck squamous cell carcinoma, urethral cancer, gastric cancer |
| Atezolizumab | PD-L1 | Roche | IgG1 | IMpower-110, IMpower-010 | 51 | 6 | US [2016], China [2019] | Advanced bladder cancer, locally advanced or metastatic urethral cancer, Metastatic non-small cell lung cancer, locally advanced or metastatic urethral cancer |
IgG, immunoglobulin G; PD-1, programmed death receptor 1; PD-L1, programmed death ligand 1; R&D, research and development.
Nivolumab
Nivolumab is a human immunoglobulin (IgG4) anti-PD-1 monoclonal targeted drug approved by the US Food and Drug Administration (FDA) for the treatment of lung cancer, which is mainly used for single chemotherapy or combined with other chemotherapy drugs (26). However, nivolumab is no longer considered a primary option in first-line therapy for non-small cell lung cancer (NSCLC). Recent treatment guidelines now favor pembrolizumab and atezolizumab as first-line agents, either as monotherapy or in combination with chemotherapy, based on PD-L1 expression levels and tumor histology. Nivolumab is currently available as a second-line treatment for advanced lung cancer after the failure of standard platinum-based first-line chemotherapy. A phase I clinical study evaluated the efficacy and safety of nivolumab in 296 patients with advanced solid tumors, and the results showed that the objective response rate (ORR) of nivolumab-treated NSCLC patients was 17.1%, 1-, 2- and 3-year survival rates were 42%, 24%, and 18%, respectively (26). Horn et al. randomized 582 patients to nivolumab or docetaxel group, and results showed that overall survival (OS) was improved with nivolumab compared with docetaxel, with an OS of 12.2 months in the nivolumab group and 9.4 months in the docetaxel group (27). Brahmer et al. randomly selected 272 patients with lung squamous cell carcinomas (LUSC) who failed platinum-based first-line chemotherapy and divided them into two groups: one group received nivolumab at a dose of 3 mg per kilogram of body weight every 2 weeks, and the other one received docetaxel, at a dose of 75 mg per square meter of body-surface area every 3 weeks. Results showed that compared with docetaxel, median survival was longer with nivolumab (9.2 vs. 6 months) and a higher ORR (20% vs. 9%). In terms of safety, nivolumab had fewer grade 3 or 4 chemotherapy-related adverse events than docetaxel (7% vs. 55%) (28). Based on the data from the above clinical studies, in March 2015, the FDA approved nivolumab for the use of platinum-containing doublet chemotherapy in patients with LUSC, and in October of the same year, it was approved for the second-line treatment of non-squamous NSCLC. As such, nivolumab’s role in lung cancer has become more historically relevant or confined to second-line settings. For a detailed overview of current first-line immunotherapy strategies, including KEYNOTE-189 and IMpower110.
Pembrolizumab
Pembrolizumab, a monoclonal antibody to human immunoglobulin (IgG4) against PD-1, is used as first-line therapy for advanced lung cancer with PD-L1 expression ≥50% and second-line therapy for PD-L1 expression ≥1%. In addition to its use in monotherapy settings, pembrolizumab has been established as a standard first-line treatment in combination with chemotherapy, based on two landmark clinical trials: KEYNOTE-189 for non-squamous NSCLC and KEYNOTE-407 for squamous NSCLC. A total of 495 patients with advanced lung cancer were included in the pembrolizumab phase I clinical study, and ultimately, both the initial and retreatment patients could achieve an effective response rate (24.8% vs. 18.0%) (29). Herbst et al. randomly assigned 1,034 patients with NSCLC (all histological types) and PD-L1 expression of at least 1% to pembrolizumab or docetaxel. The results showed that patients in the two groups who received pembrolizumab had better survival, with OS of 10.4 and 12.7 months, respectively, compared with 8.5 months for docetaxel. Moreover, patients with PD-L1 expression higher than 50% had an OS of 17.3 months, suggesting a correlation between PD-L1 expression and the efficacy of pembrolizumab (30). In KEYNOTE-189, the combination of pembrolizumab with pemetrexed and platinum chemotherapy significantly improved OS [22.0 vs. 10.7 months; hazard ratio (HR), 0.49; P<0.001] in patients with non-squamous NSCLC regardless of PD-L1 status (31). Similarly, the KEYNOTE-407 trial showed survival benefit in patients with squamous NSCLC treated with pembrolizumab plus chemotherapy (15.9 vs. 11.3 months; HR, 0.64; P=0.001) (32). Reck et al. presented the results of the phase III KEYNOTE 024 study at the European Society of Oncology Congress, which showed that pembrolizumab was more effective than platinum-based chemotherapy in the initial treatment of advanced NSCLC with high PD-L1 expression (>50%). A total of 305 patients were randomly assigned 1:1 to pembrolizumab and chemotherapy, and results revealed that the progression-free survival (PFS) of pembrolizumab and chemotherapy were 10.3 and 6.0 months, respectively, and the ORR of pembrolizumab and chemotherapy were 45% and 8%, respectively, with fewer adverse events than chemotherapy (27% vs. 53%) (33). The 2017 National Comprehensive Cancer Network (NCCN) guidelines mention first-line use of pembrolizumab in patients with PD-L1 expression greater than 50% and no driver mutations (34). More recently, KEYNOTE-671 demonstrated that perioperative pembrolizumab (neoadjuvant plus adjuvant) significantly improved event-free survival and pathological complete response in patients with resectable stage II–IIIB NSCLC, suggesting its utility even in early-stage disease (35).
Atezolizumab
Atezolizumab, a monoclonal antibody targeting programmed death ligand (PD-L1 or CD274 antigen), was developed by Genentech for the treatment of various hematological malignancies and solid tumors. It has been approved as a second-line treatment for transitional cell carcinoma in the United States and is undergoing clinical trials as a second-line treatment for cell lung cancer. In a phase I study to evaluate atezolizumab monotherapy in patients with different types of advanced tumors, 53 patients in the NSCLC group were evaluable, with a response rate of 23% and a median PFS of 15 weeks. Mazieres et al. conducted an open randomized controlled trial of patients with advanced lung cancer who had progressed after first-line chemotherapy, in which 287 patients were enrolled in a random group to receive atezolizumab or docetaxel. Results showed that the OS was longer in the atezolizumab group than in the docetaxel group (12.6 vs. 9.7 months), indicating that atezolizumab significantly improved OS in the total population (36). In the IMpower110 trial, atezolizumab was evaluated as a first-line monotherapy in patients with stage IV NSCLC expressing PD-L1 on ≥1% of tumor cells or immune cells. In patients with high PD-L1 expression (TC3 or IC3), atezolizumab significantly prolonged OS compared to chemotherapy, with a median OS of 20.2 vs. 13.1 months (HR, 0.59; 95% confidence interval: 0.40–0.89; P=0.01) (37). This study led to the approval of atezolizumab as a first-line option in PD-L1-high patients, particularly those not suitable for combination chemo-immunotherapy regimens.
Adverse reactions of PD-1/PD-L1 inhibitors in lung cancer
Immune checkpoint inhibitors prevent tumor immune escape and enhance the body’s anti-tumor effect by blocking the negative regulatory signals of T cells, but at the same time, they can also enhance the normal immune response of the body, resulting in excessive activation of the immune system and triggering an autoimmune response, that is, the immune-related adverse events (irAEs) (38). Among endocrine-related irAEs, thyroid dysfunction—particularly thyroiditis and hypothyroidism—is one of the most common, occurring in approximately 10–20% of patients. Thyroiditis often begins as transient hyperthyroidism and progresses to hypothyroidism, which may require long-term thyroid hormone replacement therapy. Routine monitoring of thyroid function during immunotherapy is recommended to enable early detection and intervention. With the wide application of immune checkpoint inhibitors, irAEs are also increasing, which often occur in the early stage after the start of treatment. The most common adverse reactions include skin diseases, gastrointestinal tract, endocrine, and pulmonary, cardiovascular, kidney or nervous system, etc. (39). A study has shown that only 5% of lung cancer patients who received anti-PD-1/PD-L1 immunotherapy in phase I clinical studies had irAEs, including fatigue, gastrointestinal discomfort, and skin reactions, among which grade 3–4 irAEs accounted for 14% (40). Later phase II clinical trials (CheckMate 063) and large randomized phase III controlled studies (CheckMate 017) found similar AEs: fatigue, anorexia, nausea, asthenia, rash, diarrhea, etc.; the most common grade 3–4 AEs were mainly fatigue, anorexia, diarrhea, and pneumonia. Therefore, in the course of immunotherapy, doctors need to fully understand potential adverse reactions and closely monitor patients. If adverse reactions occur, treatment should be implemented as soon as possible, and preventing patients from organ dysfunction and death is the key to successful treatment.
The management of irAEs depends on their severity, which is assessed using the Common Terminology Criteria for Adverse Events (CTCAE, version 5.0) (41). Grade 1 (mild) events generally allow continuation of immunotherapy with close observation. In contrast, grade 2 (moderate) events often require temporary suspension of immune checkpoint inhibitors and the initiation of corticosteroids, such as prednisone at a dose of 0.5–1 mg/kg/day. For grade 3 or higher (severe) toxicities, permanent discontinuation of immunotherapy is typically necessary, along with the administration of high-dose corticosteroids (1–2 mg/kg/day). In steroid-refractory cases—particularly with colitis or pneumonitis—additional immunosuppressive agents such as infliximab may be indicated. Prompt identification and early intervention are essential to reducing morbidity, preventing irreversible organ damage, and maintaining the overall continuity of cancer immunotherapy.
Discussion and outlook
Over the past decade, PD-1/PD-L1 immune checkpoint inhibitors have transitioned from experimental second-line treatments to well-established first-line therapies for NSCLC. Numerous large-scale clinical trials—such as KEYNOTE-189, KEYNOTE-407, and IMpower110—have validated their efficacy in prolonging survival across diverse patient populations, and these agents are now central to frontline treatment strategies (31-33,37). Mechanistically, PD-1/PD-L1 inhibitors restore anti-tumor T cell activity by relieving inhibitory signaling in the TME, thereby reversing immune suppression and reinvigorating effector responses (42,43).
Despite their success, immune checkpoint inhibitors are not universally effective. A substantial proportion of patients fail to respond initially (primary resistance) or relapse after an initial benefit (acquired resistance). These failures are driven by complex mechanisms, including loss of antigen presentation, alterations in oncogenic pathways, expansion of immunosuppressive cells (e.g., Tregs, MDSCs), and T cell exhaustion (44,45). Consequently, a critical challenge in the field is to overcome such resistance and improve long-term outcomes.
To that end, several next-generation immunotherapeutic strategies are being explored. Bispecific antibodies targeting both PD-L1 and other suppressive mediators, such as TGF-β, offer the ability to modulate multiple resistance pathways simultaneously. Furthermore, emerging immune checkpoints such as TIGIT and LAG-3 have gained traction as targets for combinatorial regimens. For example, tiragolumab, a TIGIT inhibitor, has shown promising results when combined with atezolizumab in PD-L1-positive NSCLC patients in the CITYSCAPE trial and ongoing SKYSCRAPER-01 study (46). Similarly, relatlimab, a LAG-3-blocking antibody, demonstrated improved PFS when combined with nivolumab in the RELATIVITY-047 study in melanoma, and is under investigation in lung cancer (47). A mechanistic study further supports LAG-3’s role in promoting T cell dysfunction, making it a compelling co-target with PD-1 blockade (48).
Looking ahead, the continued evolution of lung cancer immunotherapy will depend on integrating immune phenotyping, biomarker-driven patient selection, and rational combination therapy. As our understanding of the tumor-immune interface deepens, the development of tailored immunotherapeutic approaches holds promise to overcome resistance and extend durable responses to a broader range of patients.
Conclusions
In conclusion, while PD-1/PD-L1 inhibitors have revolutionized the treatment of NSCLC, several clinical and biological challenges remain. These include resistance, toxicity, and limited biomarker guidance, which restrict the scope of durable benefit. Future efforts should focus on rational combination therapies, immune phenotype–based patient selection, and integrated biomarker strategies to optimize treatment outcomes. Table 2 provides an overview of these challenges, their mechanistic basis, and potential research directions to guide the next phase of immunotherapy development.
Table 2. Immunotherapy challenges and strategies.
| Current challenges | Underlying mechanisms | Future strategies |
|---|---|---|
| Primary and acquired resistance | Loss of antigen presentation; T cell exhaustion | Dual checkpoint blockade (e.g., PD-1 + TIGIT/LAG-3) |
| Limited predictive biomarkers | PD-L1 expression alone is insufficient; TMB variability | Use of composite biomarkers (PD-L1 + TMB + TILs) |
| Immune-related adverse events (irAEs) | Systemic immune activation; autoimmunity | Biomarker-guided toxicity prediction; steroid-sparing agents |
| Incomplete tumor immune infiltration | Stromal barriers; VEGF-mediated exclusion | Anti-VEGF combination; TME remodeling |
| Lack of personalized immunotherapy | Heterogeneity in immune microenvironment | Immune phenotype-based therapy selection |
PD-1, programmed death receptor 1; PD-L1, programmed death ligand 1; TILs, tumor-infiltrating lymphocytes; TMB, tumor mutation burden; TME, tumor microenvironment; VEGF, vascular endothelial growth factor.
Supplementary
The article’s supplementary files as
Acknowledgments
None.
Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.
Footnotes
Funding: This work was supported by the Natural Science Foundation of Jilin Province (grant No. YDZJ202201ZYTS188).
Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tcr.amegroups.com/article/view/10.21037/tcr-2025-230/coif). The authors have no conflicts of interest to declare.
References
- 1.Bray F, Laversanne M, Sung H, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2024;74:229-63. 10.3322/caac.21834 [DOI] [PubMed] [Google Scholar]
- 2.Scheffler M, Michels S, Nogova L. Targeted treatment of non-small cell lung cancer. Inn Med (Heidelb) 2022;63:700-8. 10.1007/s00108-022-01372-2 [DOI] [PubMed] [Google Scholar]
- 3.Ishida Y, Agata Y, Shibahara K, et al. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J 1992;11:3887-95. 10.1002/j.1460-2075.1992.tb05481.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Lai J, Beavis PA, Li J, et al. Augmenting Adoptive T-cell Immunotherapy by Targeting the PD-1/PD-L1 Axis. Cancer Res 2021;81:5803-5. 10.1158/0008-5472.CAN-21-3548 [DOI] [PubMed] [Google Scholar]
- 5.Liu C, Zheng S, Jin R, et al. The superior efficacy of anti-PD-1/PD-L1 immunotherapy in KRAS-mutant non-small cell lung cancer that correlates with an inflammatory phenotype and increased immunogenicity. Cancer Lett 2020;470:95-105. 10.1016/j.canlet.2019.10.027 [DOI] [PubMed] [Google Scholar]
- 6.Calles A, Liao X, Sholl LM, et al. Expression of PD-1 and Its Ligands, PD-L1 and PD-L2, in Smokers and Never Smokers with KRAS-Mutant Lung Cancer. J Thorac Oncol 2015;10:1726-35. 10.1097/JTO.0000000000000687 [DOI] [PubMed] [Google Scholar]
- 7.Sznol M, Chen L. Antagonist antibodies to PD-1 and B7-H1 (PD-L1) in the treatment of advanced human cancer. Clin Cancer Res 2013;19:1021-34. 10.1158/1078-0432.CCR-12-2063 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Abate-Daga D, Hanada K, Davis JL, et al. Expression profiling of TCR-engineered T cells demonstrates overexpression of multiple inhibitory receptors in persisting lymphocytes. Blood 2013;122:1399-410. 10.1182/blood-2013-04-495531 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Liu C, Zheng S, Wang Z, et al. KRAS-G12D mutation drives immune suppression and the primary resistance of anti-PD-1/PD-L1 immunotherapy in non-small cell lung cancer. Cancer Commun (Lond) 2022;42:828-47. 10.1002/cac2.12327 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Piao W, Li L, Saxena V, et al. PD-L1 signaling selectively regulates T cell lymphatic transendothelial migration. Nat Commun 2022;13:2176. 10.1038/s41467-022-29930-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Yearley JH, Gibson C, Yu N, et al. PD-L2 Expression in Human Tumors: Relevance to Anti-PD-1 Therapy in Cancer. Clin Cancer Res 2017;23:3158-67. 10.1158/1078-0432.CCR-16-1761 [DOI] [PubMed] [Google Scholar]
- 12.David P, Megger DA, Kaiser T, et al. The PD-1/PD-L1 Pathway Affects the Expansion and Function of Cytotoxic CD8(+) T Cells During an Acute Retroviral Infection. Front Immunol 2019;10:54. 10.3389/fimmu.2019.00054 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Sun J, Zheng Y, Mamun M, et al. Research progress of PD-1/PD-L1 immunotherapy in gastrointestinal tumors. Biomed Pharmacother 2020;129:110504. 10.1016/j.biopha.2020.110504 [DOI] [PubMed] [Google Scholar]
- 14.Wang F, Yang L, Xiao M, et al. PD-L1 regulates cell proliferation and apoptosis in acute myeloid leukemia by activating PI3K-AKT signaling pathway. Sci Rep 2022;12:11444. 10.1038/s41598-022-15020-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Ma XM, Luo YF, Zeng FF, et al. TGF-β1-Mediated PD-L1 Glycosylation Contributes to Immune Escape via c-Jun/STT3A Pathway in Nasopharyngeal Carcinoma. Front Oncol 2022;12:815437. 10.3389/fonc.2022.815437 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Gao Z, Chen JF, Li XG, et al. KRAS acting through ERK signaling stabilizes PD-L1 via inhibiting autophagy pathway in intrahepatic cholangiocarcinoma. Cancer Cell Int 2022;22:128. 10.1186/s12935-022-02550-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Bie F, Tian H, Sun N, et al. Research Progress of Anti-PD-1/PD-L1 Immunotherapy Related Mechanisms and Predictive Biomarkers in NSCLC. Front Oncol 2022;12:769124. 10.3389/fonc.2022.769124 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Shurin MR, Umansky V. Cross-talk between HIF and PD-1/PD-L1 pathways in carcinogenesis and therapy. J Clin Invest 2022;132:e159473. 10.1172/JCI159473 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Barsoum IB, Smallwood CA, Siemens DR, et al. A mechanism of hypoxia-mediated escape from adaptive immunity in cancer cells. Cancer Res 2014;74:665-74. 10.1158/0008-5472.CAN-13-0992 [DOI] [PubMed] [Google Scholar]
- 20.Cheng B, Ding K, Chen P, et al. Anti-PD-L1/TGF-betaR fusion protein (SHR-1701) overcomes disrupted lymphocyte recovery-induced resistance to PD-1/PD-L1 inhibitors in lung cancer. Cancer Commun (Lond) 2022;42:17-36. 10.1002/cac2.12244 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Stephen TL, Rutkowski MR, Allegrezza MJ, et al. Transforming growth factor β-mediated suppression of antitumor T cells requires FoxP1 transcription factor expression. Immunity 2014;41:427-39. 10.1016/j.immuni.2014.08.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Stephen TL, Payne KK, Chaurio RA, et al. SATB1 Expression Governs Epigenetic Repression of PD-1 in Tumor-Reactive T Cells. Immunity. 2017;46:51-64. 10.1016/j.immuni.2016.12.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Allen SD, Liu X, Jiang J, et al. Immune checkpoint inhibition in syngeneic mouse cancer models by a silicasome nanocarrier delivering a GSK3 inhibitor. Biomaterials 2021;269:120635. 10.1016/j.biomaterials.2020.120635 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Taylor A, Harker JA, Chanthong K, et al. Glycogen Synthase Kinase 3 Inactivation Drives T-bet-Mediated Downregulation of Co-receptor PD-1 to Enhance CD8(+) Cytolytic T Cell Responses. Immunity 2016;44:274-86. 10.1016/j.immuni.2016.01.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Taylor A, Rudd CE. Small Molecule Inhibition of Glycogen Synthase Kinase-3 in Cancer Immunotherapy. Adv Exp Med Biol 2019;1164:225-33. 10.1007/978-3-030-22254-3_17 [DOI] [PubMed] [Google Scholar]
- 26.Gettinger SN, Horn L, Gandhi L, et al. Overall Survival and Long-Term Safety of Nivolumab (Anti-Programmed Death 1 Antibody, BMS-936558, ONO-4538) in Patients With Previously Treated Advanced Non-Small-Cell Lung Cancer. J Clin Oncol 2015;33:2004-12. 10.1200/JCO.2014.58.3708 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Horn L, Spigel DR, Vokes EE, et al. Nivolumab Versus Docetaxel in Previously Treated Patients With Advanced Non-Small-Cell Lung Cancer: Two-Year Outcomes From Two Randomized, Open-Label, Phase III Trials (CheckMate 017 and CheckMate 057). J Clin Oncol 2017;35:3924-33. 10.1200/JCO.2017.74.3062 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Brahmer J, Reckamp KL, Baas P, et al. Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer. N Engl J Med 2015;373:123-35. 10.1056/NEJMoa1504627 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Zhu L, Jing S, Wang B, et al. Anti-PD-1/PD-L1 Therapy as a Promising Option for Non-Small Cell Lung Cancer: a Single arm Meta-Analysis. Pathol Oncol Res 2016;22:331-9. 10.1007/s12253-015-0011-z [DOI] [PubMed] [Google Scholar]
- 30.Herbst RS, Baas P, Kim DW, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet 2016;387:1540-50. 10.1016/S0140-6736(15)01281-7 [DOI] [PubMed] [Google Scholar]
- 31.Gandhi L, Rodríguez-Abreu D, Gadgeel S, et al. Pembrolizumab plus Chemotherapy in Metastatic Non-Small-Cell Lung Cancer. N Engl J Med 2018;378:2078-92. 10.1056/NEJMoa1801005 [DOI] [PubMed] [Google Scholar]
- 32.Paz-Ares L, Luft A, Vicente D, et al. Pembrolizumab plus Chemotherapy for Squamous Non-Small-Cell Lung Cancer. N Engl J Med 2018;379:2040-51. 10.1056/NEJMoa1810865 [DOI] [PubMed] [Google Scholar]
- 33.Reck M, Rodríguez-Abreu D, Robinson AG, et al. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N Engl J Med 2016;375:1823-33. 10.1056/NEJMoa1606774 [DOI] [PubMed] [Google Scholar]
- 34.Ettinger DS, Wood DE, Aggarwal C, et al. NCCN Guidelines Insights: Non-Small Cell Lung Cancer, Version 1.2020. J Natl Compr Canc Netw 2019;17:1464-72. 10.6004/jnccn.2019.0059 [DOI] [PubMed] [Google Scholar]
- 35.Wakelee H, Liberman M, Kato T, et al. Perioperative Pembrolizumab for Early-Stage Non-Small-Cell Lung Cancer. N Engl J Med 2023;389:491-503. 10.1056/NEJMoa2302983 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Mazieres J, Rittmeyer A, Gadgeel S, et al. Atezolizumab Versus Docetaxel in Pretreated Patients With NSCLC: Final Results From the Randomized Phase 2 POPLAR and Phase 3 OAK Clinical Trials. J Thorac Oncol 2021;16:140-50. 10.1016/j.jtho.2020.09.022 [DOI] [PubMed] [Google Scholar]
- 37.Herbst RS, Giaccone G, de Marinis F, et al. Atezolizumab for First-Line Treatment of PD-L1-Selected Patients with NSCLC. N Engl J Med 2020;383:1328-39. 10.1056/NEJMoa1917346 [DOI] [PubMed] [Google Scholar]
- 38.Bhardwaj M, Chiu MN, Pilkhwal Sah S. Adverse cutaneous toxicities by PD-1/PD-L1 immune checkpoint inhibitors: pathogenesis, treatment, and surveillance. Cutan Ocul Toxicol 2022;41:73-90. 10.1080/15569527.2022.2034842 [DOI] [PubMed] [Google Scholar]
- 39.Teufel A, Zhan T, Härtel N, et al. Management of immune related adverse events induced by immune checkpoint inhibition. Cancer Lett 2019;456:80-7. 10.1016/j.canlet.2019.04.018 [DOI] [PubMed] [Google Scholar]
- 40.Brahmer JR, Hammers H, Lipson EJ. Nivolumab: targeting PD-1 to bolster antitumor immunity. Future Oncol 2015;11:1307-26. 10.2217/fon.15.52 [DOI] [PubMed] [Google Scholar]
- 41.Johnson M, Dudek AZ, Sukari A, et al. ARRY-382 in Combination with Pembrolizumab in Patients with Advanced Solid Tumors: Results from a Phase 1b/2 Study. Clin Cancer Res 2022;28:2517-26. 10.1158/1078-0432.CCR-21-3009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Nagaraju GP, Malla RR, Basha R, et al. Contemporary clinical trials in pancreatic cancer immunotherapy targeting PD-1 and PD-L1. Semin Cancer Biol 2022;86:616-21. 10.1016/j.semcancer.2021.11.003 [DOI] [PubMed] [Google Scholar]
- 43.Girault I, Adam J, Shen S, et al. A PD-1/PD-L1 Proximity Assay as a Theranostic Marker for PD-1 Blockade in Patients with Metastatic Melanoma. Clin Cancer Res 2022;28:518-25. 10.1158/1078-0432.CCR-21-1229 [DOI] [PubMed] [Google Scholar]
- 44.Gavrielatou N, Liu Y, Vathiotis I, et al. Association of PD-1/PD-L1 Co-location with Immunotherapy Outcomes in Non-Small Cell Lung Cancer. Clin Cancer Res 2022;28:360-7. 10.1158/1078-0432.CCR-21-2649 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Lee DY, Im E, Yoon D, et al. Pivotal role of PD-1/PD-L1 immune checkpoints in immune escape and cancer progression: Their interplay with platelets and FOXP3+Tregs related molecules, clinical implications and combinational potential with phytochemicals. Semin Cancer Biol 2022;86:1033-57. 10.1016/j.semcancer.2020.12.001 [DOI] [PubMed] [Google Scholar]
- 46.Cho BC, Abreu DR, Hussein M, et al. Tiragolumab plus atezolizumab versus placebo plus atezolizumab as a first-line treatment for PD-L1-selected non-small-cell lung cancer (CITYSCAPE): primary and follow-up analyses of a randomised, double-blind, phase 2 study. Lancet Oncol 2022;23:781-92. 10.1016/S1470-2045(22)00226-1 [DOI] [PubMed] [Google Scholar]
- 47.Tawbi HA, Schadendorf D, Lipson EJ, et al. Relatlimab and Nivolumab versus Nivolumab in Untreated Advanced Melanoma. N Engl J Med 2022;386:24-34. 10.1056/NEJMoa2109970 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Andrews LP, Yano H, Vignali DAA. Inhibitory receptors and ligands beyond PD-1, PD-L1 and CTLA-4: breakthroughs or backups. Nat Immunol 2019;20:1425-34. 10.1038/s41590-019-0512-0 [DOI] [PubMed] [Google Scholar]


