Skip to main content

MINI REVIEW article

Front. Immunol., 12 December 2024
Sec. Cancer Immunity and Immunotherapy
This article is part of the Research Topic Dissecting the Role of T Cell Exhaustion in Cancer Progression: A Multifaceted Approach View all 3 articles

Targeting T cell exhaustion: emerging strategies in non-small cell lung cancer

Xianqiang Liu,&#x;&#x;Xianqiang Liu1,2†‡Xiaowei Xi&#x;Xiaowei Xi3†Shengshan Xu*&#x;Shengshan Xu1*†Hongyu Chu&#x;Hongyu Chu4†Penghui HuPenghui Hu5Dong LiDong Li6Bin Zhang,Bin Zhang7,8Hejie LiuHejie Liu1Tianxiao Jiang*Tianxiao Jiang9*Zhuming Lu*Zhuming Lu1*
  • 1Department of Thoracic Surgery, Jiangmen Central Hospital, Jiangmen, Guangdong, China
  • 2Graduate School, Medical School of Chinese PLA, Beijing, China
  • 3Technical University of Munich (TUM) School of Medicine and Health, Munich, Germany
  • 4Department of Gastrointestinal, Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
  • 5Scientific Research and Education Department, Jiangmen Central Hospital, Jiangmen, Guangdong, China
  • 6Department of Intensive Care Unit and Clinical Experimental Center, Jiangmen Central Hospital, Jiangmen, China
  • 7Department of Cardiovascular Disease and Clinical Experimental Center, Jiangmen Central Hospital, Jiangmen, China
  • 8Department of Cardiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
  • 9Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany

Lung cancer continues to be a major contributor to cancer-related deaths globally. Recent advances in immunotherapy have introduced promising treatments targeting T cell functionality. Central to the efficacy of these therapies is the role of T cells, which are often rendered dysfunctional due to continuous antigenic stimulation in the tumor microenvironment–a condition referred to as T cell exhaustion. This review addresses the critical challenge of T cell exhaustion in non-small cell lung cancer (NSCLC), offering a detailed examination of its molecular underpinnings and the resultant therapeutic ineffectiveness. We synthesize current knowledge on the drivers of T cell exhaustion, evaluate emerging strategies for its reversal, and explore the potential impact of these insights for enhancing the clinical efficacy of immunotherapies. By consolidating reported clinical trials and preclinical studies, this article highlights innovative approaches to modulate immune responses and improve patient outcomes, thus providing a roadmap for future research and therapeutic development in lung cancer immunotherapy.

1 Introduction

Lung cancer continues to pose a significant global health issue, consistently ranking as the second most diagnosed malignancy as of 2020 and the leading contributor to cancer-related mortality. This disease is primarily divided into two subtypes: small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC) (1). SCLC, representing about 15% of all lung cancer instances, characterized by high aggressiveness and low survival rates, with a five-year survival rate of 7% (2). NSCLC comprises about 85% of cases and has a somewhat higher five-year survival rate of around 25% (3). Despite advancements in treatment, about half of all lung cancer patients are diagnosed with advanced stages of the disease, undergoing treatment regimens predominantly based on platinum-based chemotherapy (4). Even with the integration of chemotherapy and targeted therapies or other treatment modalities, the average five-year survival rate languishes at approximately 6%, underscoring a dire need for more effective therapeutic options (5).

In recent years, the emergence of immunotherapy has diversified the treatment modalities for lung cancer. T cells are vital for the effectiveness of immunotherapies, but their function can be significantly hindered by continuous antigen exposure from chronic infections or tumors, leading to T cell exhaustion. This state is marked by diminished production of effector cytokines such as IL-2 and IFN-γ, reduced proliferative capacity, and compromised cytotoxic and pro-inflammatory functions (68). The exacerbated expression of multiple immune checkpoints like PD-1, CTLA-4, LAG-3, TIGIT, BTLA, and TIM-3 further illustrates the impaired functionality of T cells under such conditions (9, 10). Consequently, many patients receiving immunotherapies like PD-1 or PD-L1 inhibitors do not maintain strong anti-tumor effects (11), propelling ongoing research to focus on methods to reverse T cell exhaustion (12). This review seeks to delve into emerging strategies to rejuvenate T cell activity, potentially counteracting the negative impact of continuous antigenic stimulation and fostering a more effective pathway for lung cancer treatment.

2 Inhibitory receptors involved in T cell exhaustion in lung cancer

Elevated expression of multiple inhibitory receptors has been well-documented in lung cancer patients, signifying a state of T cell exhaustion (13). Further complicating the treatment landscape, resistance to PD-1 inhibitors in lung cancer has been correlated with upregulation of other immune checkpoints such as TIM-3 and LAG-3 in mouse models (14).

2.1 PD-1

Programmed death receptor-1 (PD-1), a member of the CD28 transmembrane protein receptor family, plays a critical role in the immune system’s ability to respond to tumor cells. PD-1 suppresses the anti-cancer immune activity of T cells through its interaction with ligands PD-L1 or PD-L2 (15, 16), leading to downregulation of the T cell receptor and subsequent inhibition of T cell activation and cytokine release (17, 18). Notably, interferon-gamma activates the PI3K/AKT and JAK/STAT3 pathways in NSCLC, which enhances PD-L1 expression, a mechanism indicating IFN-γ induced immune suppression (19). Innovative treatments, such as vaccines using plasmacytoid dendritic cells in conjunction with anti-PD-1 therapies, have proved promise in amplifying the tumor-specific CD8+ T cell response in lung cancer (20). Other studies indicate that the miR-3127-5p/p-STAT3 axis (21), the EZH2-HIF-1α axis (22), KLF12 (23), and POU2F1 (24) are also involved in controlling PD-L1 expression, ultimately affecting T cell exhaustion. Therefore, a deeper understanding of the mechanisms that regulate PD-L1 expression could lead to improved strategies for enhancing the effectiveness of PD-L1/PD-1 inhibitors in lung cancer treatment. For a detailed summary of clinical trials employing PD-1 inhibitors in lung cancer, refer to Supplementary Table 1, which catalogues and summarizes these trials.

2.2 CTLA-4

As another inhibitory member of the CD28 family, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), mainly found on T cells, CTLA-4 competitively binds with the ligands CD80/CD86, curtailing CD28 signaling and subsequently reducing IL-2 production and T cell-APC contact time-crucially weakening T cell responses (25). This receptor also triggers the PI3-K pathway, diminishing T cell effectiveness further. The enhancement of PD-1/PD-L1 and CTLA-4 expression via the EGFR pathway suggests a potent avenue for immune suppression in lung cancer, exacerbated by ERK and NF-κB pathways (26). In a Phase Ib/II study (NCT04646330), researchers assessed the combined use of cadonilimab (a PD-1/CTLA-4 bispecific antibody) with anlotinib as a primary treatment for advanced NSCLC, revealing the regimen’s safety and potential efficacy at 10 mg/kg every three weeks. Employing anti-PD-1/PD-L1 and anti-CTLA-4 drugs targets different stages of the cancer immunity cycle; their combined use can produce a synergistic effect, helping to overcome resistance to monotherapy (27, 28). Additionally, Supplementary Table 2 provides an overview of clinical trials investigating CTLA-4 inhibitors in lung cancer.

2.3 TIM-3

T cell immunoglobulin and mucin-domain containing-3 (TIM-3), a member of the immunoglobulin superfamily, was first identified on CD4+ Th1 and CD8+ Tc1 cells as a surface marker that binds to galectin-9. This interaction negatively regulates the function of these cells (29, 30). TIM-3 is also found on a variety of cells within the tumor microenvironment (TME), such as NK cells, dendritic cells (DCs), and tumor cells, which underscores its pivotal role in disease progression in lung cancer (30, 31). High levels of TIM-3 on NK cells and tumor-associated macrophages are related to advanced disease stages and reduced survival rates in lung cancer patients (32, 33). Therapeutically, combining anti-TIM-3 with anti-PD-1 therapies has been effective in reviving the effector functions of CD8 T cells in vitro, indicating that TIM-3 plays a significant role not only as a biomarker of resistance but also in the mechanisms of resistance itself. A Phase I/II trial (NCT02608268) evaluating MGB453 (an anti-TIM-3 drug) combined with PDR001 (an anti-PD-1 therapy) demonstrated good safety and antitumor activity, highlighting the clinical potential of targeting TIM-3 in advanced NSCLC (34). Ongoing clinical trials continue to assess the efficacy of several novel TIM-3 inhibitors, including AZD7789, INCAGN02390, BMS-986258, cobolimab, and sabatolimab (MBG453) (35).

2.4 LAG3

Lymphocyte activation gene-3 (LAG-3) exhibits structural similarities to CD4 in its extracellular domain, yet its intracellular domain markedly differs, indicating unique functional attributes (36). LAG-3 more strongly binds MHC-II compared to CD4, effectively interfering with CD4’s interaction with MHC-II, ultimately suppressing T cell activation (37). Unlike naïve T cells, LAG-3 is expressed on CD4+ and CD8+ T cells after antigen exposure, serving to inhibit their proliferation (38, 39). In the TME, constant antigen exposure heightens LAG-3 expression, contributing to T cell exhaustion and impaired tumor cell elimination. T cells that express both LAG-3 and PD-1 exhibit a greater degree of exhaustion than those expressing PD-1 alone, making LAG-3 a potential target for rescuing dysfunctional T cells in lung cancer. A Phase II study (NCT03365791) evaluating LAG525 combined with PDR001 in advanced solid tumors showed promising anti-tumor effects, achieving a 24-week clinical benefit rate of 27% in SCLC cases. Additionally, Xencor’s bispecific antibody XmAb®22841 targets both CTLA-4 and LAG-3, boosting T cell activation and proliferation (40). A Phase I study (NCT03849469) is currently exploring the optimal dose of XmAb22841, alone or with pembrolizumab, for treating advanced solid tumors, including SCLC (41).

2.5 TIGIT

The T cell immunoreceptor with Ig and ITIM domains (TIGIT) is a recently identified immune checkpoint receptor primarily found on T cells and NK cells, binding to the ligand CD155. TIGIT serves as a marker for T cell exhaustion, effectively identifying exhausted T cells across different stages of differentiation and fostering the exhaustion of CD8 T cells and NK cells under chronic conditions (4244). Furthermore, research by Yang et al. found that the positivity rate of CD155 in the immunohistochemistry results of squamous lung cancer is significantly higher than that of PD-L1, indicating a crucial role for the TIGIT/CD155 axis in the development and progression of this cancer type (45). Additionally, high TIGIT levels are linked to greater severity in lung adenocarcinoma, and patients with overexpressed CD155 in lung adenocarcinoma and SCLC have shorter progression-free survival and overall survival (4648). Studies show that antagonistic antibodies against TIGIT, when used with PD-1 inhibitors, effectively curb lung cancer growth in immunocompetent mice (49). Ongoing clinical trials, such as NCT02964013 (MK-7684-001), are assessing the efficacy of the anti-TIGIT antibody vibostolimab in combination with pembrolizumab, demonstrating promising antitumor effects particularly in patients new to anti-PD-1/PD-L1 therapy and those with PD-L1 positivity (50).

2.6 BTLA

The B and T lymphocyte attenuator (BTLA) belongs to the immunoglobulin superfamily and is primarily found on the surfaces of T cells, B cells, NK cells, and DCs (51, 52). As an inhibitory receptor, BTLA limits the activation, proliferation, and production of pro-inflammatory cytokines (IFN-γ, IL-2, and IL-10) in T and B cells through its interaction with the ligand HVEM (53). In lung cancer, BTLA plays a particularly important role, co-participating in T cell exhaustion with other inhibitory receptors like PD-1 and CTLA-4 (54, 55). Within the TME, elevated BTLA levels result in reduced T cell efficacy, affecting their immune response to tumors (56). Studies by Mittal et al. and Thommen et al. demonstrate that BTLA, along with markers like PD-1 and TIM-3, is upregulated in T cells post-tumor implantation in mice and in infiltrating CD8 T cells in advanced lung cancer patients, underscoring its role in T cell dysfunction during tumor progression (57, 58). Clinical studies suggest that high expression of BTLA is linked to unfavorable outcomes in NSCLC and is viewed as a new target for immunotherapy, potentially by combining other therapeutic strategies (such as PD-1 blockade) to restore T cell function (59).

2.7 IDO

Indoleamine 2,3-dioxygenase (IDO) is an intracellular enzyme that catalyzes the conversion of the essential amino acid L-tryptophan into N-formylkynurenine (60). By diminishing local tryptophan levels and augmenting the production of immunoregulatory metabolites, IDO exhibits its immunosuppressive effects (61). These metabolites not only inhibit T lymphocyte proliferation but also promote apoptosis and drive the transformation of naïve T cells into regulatory T cells. Additionally, overexpression of IDO in DCs affects their maturation, reducing antigen presentation and decreasing co-stimulatory molecule expression (60). Increasing evidence links IDO overexpression with adverse outcomes in several cancers (62, 63). IDO is considered a mechanism of resistance that might hinder the effectiveness of checkpoint inhibitor therapies. In a mouse model of lung cancer, silencing IDO1 inhibited tumor growth by reversing T cell exhaustion. Furthermore, blocking the T cell exhaustion induced by IDO1 can enhance the performance of PD-1 inhibitors in lung cancer treatment (64). Therefore, IDO as a potential immunosuppressant is worth further exploration. Table 1 provides a comprehensive overview of the clinical trials involving immune checkpoint modulators, such as TIM-3, LAG-3, TIGIT, BTLA, and IDO, in the context of lung cancer.

Table 1
www.frontiersin.org

Table 1. Clinical trials of immune checkpoint modulators (TIM3, LAG-3, TIGIT, BTLA, and IDO) in lung cancer.

3 The impact of cytokines on T cell exhaustion in lung cancer

3.1 IL-10

IL-10, a key anti-inflammatory cytokine, is secreted by various immune cells, such as DCs, B cells, CD8 T cells, and non-regulatory CD4 T cells. It plays a critical role in maintaining self-tolerance and protecting against tissue damage in inflamed conditions by inhibiting Th17 cell-mediated inflammation and reducing levels of TNF-α and IL-6 (65, 66). IL-10 exhibits a complex, dual role in antitumor immunity, with its effects shaped by various factors, including cancer type, cell type, TME conditions, and IL-10 concentration. In models of breast cancer, cutaneous squamous cell carcinoma, and lymphoma, IL-10 deficiency correlates with increased tumor burden and impaired immune surveillance, along with elevated rates of CD8+ T cell exhaustion (67, 68). Conversely, in colorectal cancer, bladder cancer, and melanoma models, IL-10 has been shown to promote CD8+ T cell exhaustion, thereby weakening antitumor immunity (6972). Additionally, in non-small cell lung cancer, IL-10, in conjunction with Tregs and IL-35, is believed to further exacerbate T cell exhaustion, suppressing antitumor responses (70).

The concentration of IL-10 plays a critical role in determining its antitumor effects. High levels of IL-10 can stimulate CD8+ T cell proliferation and enhance cytotoxicity, while low levels tend to exert immunosuppressive effects, diminishing the antitumor immune response (68). In chronic inflammatory settings, IL-10’s anti-inflammatory properties may help maintain CD8+ T cell antitumor functions (73, 74). Furthermore, IL-10 influences CD8+ T cell functionality through metabolic reprogramming, particularly by inducing oxidative phosphorylation (OXPHOS) pathways (75, 76). This upregulation of OXPHOS can partially reinvigorate exhausted T cells, enhancing their proliferative capacity, effector functions, and overall antitumor immunity (76). IL-10’s effects also vary among different cell types. For example, in B cells, IL-10 promotes proliferation, stimulates immunoglobulin secretion and isotype switching, and enhances tumor-killing capabilities (77, 78). Future research should focus on the role of IL-10 in relation to exhausted CD8+ T cells in non-small cell lung cancer, as well as the key factors influencing this dynamic, to improve the efficacy of lung cancer immunotherapies.

3.2 IFNα/β

IFNα/β are pro-inflammatory cytokines with various anti-tumor activities, such as direct tumor cell eradication and the stimulation of immune cells like DCs and CD8 T cells (7981). Studies suggest that IFNα/β can trigger the expression of IL-10, PD-L1, and other inhibitory regulators, potentially leading to T cell exhaustion through the influence on the transcription factor Tcf1 (82, 83). To date, IFNα/β has been approved for treating multiple malignancies, such as renal cell carcinoma and melanoma. Overall, the efficacy of combining PD-1/PD-L1 inhibitors with IFNα/β therapy is influenced by various factors, including the type and dosage of IFNα/β, timing and duration of treatment, patient immune status, and cancer type. Thus, understanding the mechanisms of action of IFNα/β and its interaction with PD-1/PD-L1 combined therapy is crucial for optimizing cancer treatment strategies.

3.3 TGF-β

TGF-β, a versatile cytokine, plays a critical role in regulating cell growth and differentiation. It also fosters T cell exhaustion; notably, TGF-β’s stimulation of exhausted T cell precursors leads to the inhibition of mTOR signaling, which in turn produces inhibitory cytokines that dampen the immune response (84). Furthermore, TGF-β reduces or suppresses immune cell activation through triggering SMAD transcription regulators downstream. Tumor cells release TGF-β, which directly increases PD-1 transcription in these cells, contributing to T cell exhaustion. Consequently, blocking TGF-β signaling could directly bolster anti-tumor immunity (85). The significant link between TGF-β signaling and impaired immune responses in lung cancer underscores the need to focus on this pathway when developing new therapeutic strategies. Extensive research is required to unravel its full mechanisms and evaluate the effectiveness of TGF-β inhibitors in treating lung cancer.

3.4 IL-2

IL-2, commonly referred to as T cell growth factor, is integral to T cell proliferation. At initial tumor growth stages, IL-2-driven BLIMP1 expression is vital for the development and differentiation of CD8 T cells. Recent findings suggest that IL-2 signaling can alter the differentiation trajectory of exhausted CD8 T cell precursors, potentially reversing T cell exhaustion (86). However, studies indicate that IL-2 can enhance 5-HTP production through the STAT5-TPH1 pathway, which activates the aryl hydrocarbon receptor and may lead to CD8 T cell exhaustion (87). Thus, IL-2 serves a dual function: it both contributes to and could potentially reverse the exhausted T cell phenotype. Currently, combining IL-2 with PD-1 inhibitors has demonstrated promising synergistic effects in reactivating exhausted CD8 T cells (88, 89). Ongoing research is crucial to fully ascertain the therapeutic impact of IL-2 in lung cancer treatment.

4 Non-cytokine mediators of T cell exhaustion

Beyond cytokines, non-cytokine factors such as metabolites and metabolic fuel sources like adenosine, cholesterol, and fatty acids, also contribute to T cell exhaustion (90, 91).

4.1 Prostaglandin E2

The arachidonic acid pathway, involved in immune suppression across various cancer types, includes the enzyme microsomal prostaglandin E2 synthase-1, downstream of cyclooxygenase 2, which limits the body’s anti-tumor immunity. Prostaglandin E2 (PGE2), known for its immunosuppressive effects, can suppress the activation and proliferation of immune cells such as NK cells and B cells via the PGE2-EP signaling pathway (9294). Recent research in lung cancer has shown that PGE2 promotes immune tolerance by upregulating PD-1 on infiltrating CD8 T cells, thereby contributing to their exhaustion (95). Consequently, targeting the PGE2-EP signaling pathway presents a promising approach to counteract T cell exhaustion in lung cancer therapy.

4.2 Adenosine

Adenosine (Ado) is produced from extracellular ATP through the actions of ectonucleotidases CD39 and CD73 on cell surfaces (96). Persistent high levels of Ado can contribute to an immunosuppressive microenvironment, although the precise mechanisms remain unclear (97). Ado impairs the activation of CD8 T cells in the TME and disrupts their tumor recognition capabilities, mainly through the A2AR/PKA/mTORC1 signaling pathway (98). Additionally, Ado further dampens tumor immunity by decreasing the infiltration of immune cells into tumors (99). Due to poor vascular development in tumors, the TME often exists in a hypoxic state, facilitate Ado production and activate immunosuppressive A2A and A2B receptors. Shifts in tumor metabolism towards glycolysis and the resulting increase in lactate also lower pH levels, promoting M2 polarization and inhibiting T cell activation (100, 101). Additionally, studies by Maj and colleagues have shown that during checkpoint therapy, the apoptosis of cancer cells and Tregs leads to an increase in ATP release, which is subsequently transformed into adenosine by the enzymes CD39 and CD73. The increased Ado in the TME counteracts the effects of checkpoint therapy, further suppressing anti-tumor immune responses (102). This suggests that Ado may be a potential target for advanced immunotherapies. Currently, a phase I clinical trial involving CPI-444 (an A2AR antagonist) combined with the PD-1 inhibitor nivolumab for treating non-small cell lung cancer is underway (NCT0265582).

4.3 Cholesterol

Cholesterol, a vital component of cell membranes, affects membrane fluidity as well as gene expression and metabolism, and impacting anti-tumor immunity (103). Its role in T cell activation is debated; some studies indicate it may suppress TCR signaling either by binding to the transmembrane region of TCRβ or by interfering with TCR oligomerization (104). In the TME, cholesterol is known to promote the expression of immunosuppressive receptors, leading to CD8 T cell exhaustion. Additionally, research has found that cholesterol levels in tumor-infiltrating CD8 T cells correlate with exhaustion status and the presence of immune checkpoints such as PD-1, TIM-3, and LAG-3 (104, 105). Statins, the primary compounds inhibiting cholesterol synthesis, are highly safe, with atorvastatin reported to downregulate the expression of inhibitory receptors like LAG-3, PD-1, TIM-3, and CTLA-4 in T cells, indirectly indicating the relationship between cholesterol and immune checkpoint expression (106). Moreover, statins have been found to reduce cancer-related mortality by 15% (107). Therefore, cholesterol-lowering medications may be combined with immunotherapy for cancer patients, offering new possibilities for treatment.

4.4 Clinical implications of T cell exhaustion in lung cancer immunotherapy

Understanding T cell exhaustion is crucial for monitoring and treating lung cancer. Figure 1 provides an overview of the pathways of T cell exhaustion and therapeutic interventions in lung cancer currently under study. Exhausted T cells are characterized by the expression of inhibitory receptors such as PD-1 and TIM-3, with their co-expression often signaling severe exhaustion and correlating with a poorer prognosis in lung cancer. In the TME, the presence of these markers on CD8 T cells can predict the response to immune checkpoint inhibitor therapy. Utilizing transcriptomics and multiplex staining techniques to identify patients who may benefit from ICI therapy is increasingly valuable in clinical research. Approaches to reverse T cell exhaustion and enhance anti-tumor immunity are promising, yet obstacles like immunosuppressive cytokines, certain immune cells, and elevated inhibitory receptors in the TME all play roles in promoting T cell exhaustion, which in turn dampens anti-tumor responses. Studies show that blocking inhibitory receptors and cytokines can effectively reverse T cell exhaustion in cancer patients, particularly in those with advanced disease, enhancing anti-tumor immunity. Clinical application of anti-PD-1 antibodies has been successful, and blocking PD-1 or PD-L1 improves the functionality of exhausted CD8 T cells (9, 108, 109), indicating that T cell exhaustion is not irreversible. Preclinical studies confirm that anti-PD-1 immune checkpoint inhibitors can effectively ameliorate T cell exhaustion and are more effective when combined with other immune checkpoint inhibitors (110); This has led to FDA approval for combination therapies like nivolumab plus ipilimumab (111). While single-agent CTLA-4 or PD-1 blockade has significantly extended survival in some cancer patients, most do not respond. Therefore, strategies against T cell exhaustion are becoming a focus for enhancing the effectiveness of anti-tumor therapies, especially in enhancing the efficacy and durability of treatments like CAR-T cell therapies.

Figure 1
www.frontiersin.org

Figure 1. Pathways of T cell exhaustion and therapeutic interventions in lung cancer.

5 Discussion and conclusion

Recent advances in immunotherapy have expanded treatment options for lung cancer, with T cell exhaustion emerging as a key area of interest in the disease’s pathogenesis, progression, and therapy. While the precise mechanisms behind T cell exhaustion continue to be studied, further research and clinical trials are essential to establish clear findings. Additionally, the exploration of T cell dysfunction presents a new avenue in immunotherapy. A thorough understanding of T cell exhaustion and its underlying mechanisms is critical to comprehending the immune dynamics in lung cancer. This knowledge is fundamental to crafting innovative treatment approaches for this challenging disease.

Author contributions

XL: Writing – original draft, Writing – review & editing. XX: Writing – review & editing. SX: Writing – review & editing, Writing – original draft. HC: Writing – original draft, Writing – review & editing. PH: Writing – review & editing. DL: Writing – review & editing. BZ: Writing – review & editing. HL: Writing – review & editing. TJ: Writing – review & editing. ZL: Writing – review & editing.

Funding

The author(s) declare financial support was received for the research, authorship, and/or publication of this article. This work was supported by the Technology Project of Jiangmen (Nos. 2220002000183, 2320002000933), the Medical Science Foundation of Jiangmen Central Hospital (J202401), and the Postdoctoral Project of the International Training Program for Guangdong Outstanding Young Research Talents and the General Project of China Postdoctoral Science Foundation (No. 2024M761177).

Conflict of interest

The authors declare that the research was conducted without any commercial or financial relationships that could be construed as potential conflicts of interest.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Supplementary material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fimmu.2024.1507501/full#supplementary-material

References

1. Khanmohammadi A, Aghaie A, Vahedi E, Qazvini A, Ghanei M, Afkhami A, et al. Electrochemical biosensors for the detection of lung cancer biomarkers: A review. Talanta. (2020) 206:120251. doi: 10.1016/j.talanta.2019.120251

PubMed Abstract | Crossref Full Text | Google Scholar

2. Gazdar AF, Bunn PA, Minna JD. Small-cell lung cancer: what we know, what we need to know and the path forward. Nat Rev Cancer. (2017) 17:725–37. doi: 10.1038/nrc.2017.87

PubMed Abstract | Crossref Full Text | Google Scholar

3. Arbour KC, Riely GJ. Systemic therapy for locally advanced and metastatic non-small cell lung cancer: A review. JAMA. (2019) 322:764–74. doi: 10.1001/jama.2019.11058

PubMed Abstract | Crossref Full Text | Google Scholar

4. Qu Y, Emoto K, Eguchi T, Aly RG, Zheng H, Chaft JE, et al. Pathologic assessment after neoadjuvant chemotherapy for Nsclc: importance and implications of distinguishing adenocarcinoma from squamous cell carcinoma. J Thorac Oncol. (2019) 14:482–93. doi: 10.1016/j.jtho.2018.11.017

PubMed Abstract | Crossref Full Text | Google Scholar

5. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA: A Cancer J Clin. (2021) 71:7–33. doi: 10.3322/caac.21654

PubMed Abstract | Crossref Full Text | Google Scholar

6. Pauken KE, Wherry EJ. Overcoming T cell exhaustion in infection and cancer. Trends Immunol. (2015) 36:265–76. doi: 10.1016/j.it.2015.02.008

PubMed Abstract | Crossref Full Text | Google Scholar

7. Gehring AJ, Ho ZZ, Tan AT, Aung MO, Lee KH, Tan KC, et al. Profile of tumor antigen-specific cd8 T cells in patients with hepatitis B virus-related hepatocellular carcinoma. Gastroenterology. (2009) 137:682–90. doi: 10.1053/j.gastro.2009.04.045

PubMed Abstract | Crossref Full Text | Google Scholar

8. Ahmadzadeh M, Johnson LA, Heemskerk B, Wunderlich JR, Dudley ME, White DE, et al. Tumor antigen–specific Cd8 T cells infiltrating the tumor express high levels of Pd-1 and are functionally impaired. Blood. (2009) 114:1537–44. doi: 10.1182/blood-2008-12-195792

PubMed Abstract | Crossref Full Text | Google Scholar

9. Wherry EJ. T cell exhaustion. Nat Immunol. (2011) 12:492–9. doi: 10.1038/ni.2035

PubMed Abstract | Crossref Full Text | Google Scholar

10. Franco F, Jaccard A, Romero P, Yu Y-R, Ho P-C. Metabolic and epigenetic regulation of T-cell exhaustion. Nat Metab. (2020) 2:1001–12. doi: 10.1038/s42255-020-00280-9

PubMed Abstract | Crossref Full Text | Google Scholar

11. Wang B, Zhang W, Jankovic V, Golubov J, Poon P, Oswald EM, et al. Combination cancer immunotherapy targeting Pd-1 and Gitr can rescue Cd8(+) T cell dysfunction and maintain memory phenotype. Sci Immunol. (2018) 3(29). doi: 10.1126/sciimmunol.aat7061

PubMed Abstract | Crossref Full Text | Google Scholar

12. Zhu Y, Tan H, Wang J, Zhuang H, Zhao H, Lu X. Molecular insight into T cell exhaustion in hepatocellular carcinoma. Pharmacol Res. (2024) 203:107161. doi: 10.1016/j.phrs.2024.107161

PubMed Abstract | Crossref Full Text | Google Scholar

13. Philip M, Schietinger A. Cd8+ T cell differentiation and dysfunction in cancer. Nat Rev Immunol. (2022) 22:209–23. doi: 10.1038/s41577-021-00574-3

PubMed Abstract | Crossref Full Text | Google Scholar

14. Koyama S, Akbay EA, Li YY, Herter-Sprie GS, Buczkowski KA, Richards WG, et al. Adaptive resistance to therapeutic Pd-1 blockade is associated with upregulation of alternative immune checkpoints. Nat Commun. (2016) 7:10501. doi: 10.1038/ncomms10501

PubMed Abstract | Crossref Full Text | Google Scholar

15. Dolina JS, Van Braeckel-Budimir N, Thomas GD, Salek-Ardakani S. Cd8(+) T cell exhaustion in cancer. Front Immunol. (2021) 12:715234. doi: 10.3389/fimmu.2021.715234

PubMed Abstract | Crossref Full Text | Google Scholar

16. Morad G, Helmink BA, Sharma P, Wargo JA. Hallmarks of response, resistance, and toxicity to immune checkpoint blockade. Cell. (2021) 184:5309–37. doi: 10.1016/j.cell.2021.09.020

PubMed Abstract | Crossref Full Text | Google Scholar

17. Constantinidou A, Alifieris C, Trafalis DT. Targeting programmed cell death -1 (Pd-1) and ligand (Pd-L1): A new era in cancer active immunotherapy. Pharmacol Ther. (2019) 194:84–106. doi: 10.1016/j.pharmthera.2018.09.008

PubMed Abstract | Crossref Full Text | Google Scholar

18. Dermani FK, Samadi P, Rahmani G, Kohlan AK, Najafi R. Pd-1/Pd-L1 immune checkpoint: potential target for cancer therapy. J Cell Physiol. (2019) 234:1313–25. doi: 10.1002/jcp.27172

PubMed Abstract | Crossref Full Text | Google Scholar

19. Zhang J, Liu S, Chen X, Xu X, Xu F. Non-immune cell components in tumor microenvironment influencing lung cancer immunotherapy. Biomedicine Pharmacotherapy. (2023) 166:115336. doi: 10.1016/j.biopha.2023.115336

PubMed Abstract | Crossref Full Text | Google Scholar

20. Hannani D, Leplus E, Laurin D, Caulier B, Aspord C, Madelon N, et al. A new plasmacytoid dendritic cell-based vaccine in combination with anti-Pd-1 expands the tumor-specific Cd8+ T cells of lung cancer patients. Int J Mol Sci. (2023) 24(3). doi: 10.3390/ijms24031897

PubMed Abstract | Crossref Full Text | Google Scholar

21. Tang D, Zhao D, Wu Y, Yao R, Zhou L, Lu L, et al. The Mir-3127-5p/P-Stat3 axis up-regulates Pd-L1 inducing chemoresistance in non-small-cell lung cancer. J Cell Mol Med. (2018) 22:3847–56. doi: 10.1111/jcmm.13657

PubMed Abstract | Crossref Full Text | Google Scholar

22. Zhao Y, Wang XX, Wu W, Long H, Huang J, Wang Z, et al. Ezh2 regulates Pd-L1 expression via Hif-1α in non-small cell lung cancer cells. Biochem Biophys Res Commun. (2019) 517:201–9. doi: 10.1016/j.bbrc.2019.07.039

PubMed Abstract | Crossref Full Text | Google Scholar

23. Pan X, Zhang W, Wang L, Guo H, Zheng M, Wu H, et al. Klf12 transcriptionally regulates Pd-L1 expression in non-small cell lung cancer. Mol Oncol. (2023) 17:2659–74. doi: 10.1002/1878-0261.13512

PubMed Abstract | Crossref Full Text | Google Scholar

24. Li F, Wang T, Huang Y. Pou2f1 induces the immune escape in lung cancer by up-regulating Pd-L1. Am J Trans Res. (2021) 13:672–83.

PubMed Abstract | Google Scholar

25. Rudd CE, Taylor A, Schneider H. Cd28 and Ctla-4 coreceptor expression and signal transduction. Immunol Rev. (2009) 229:12–26. doi: 10.1111/j.1600-065X.2009.00770.x

PubMed Abstract | Crossref Full Text | Google Scholar

26. Liang H, Liu X, Wang M. Immunotherapy combined with epidermal growth factor receptor-tyrosine kinase inhibitors in non-small-cell lung cancer treatment. OncoTargets Ther. (2018) 11:6189–96. doi: 10.2147/ott.S178497

PubMed Abstract | Crossref Full Text | Google Scholar

27. Feng Y, Roy A, Masson E, Chen TT, Humphrey R, Weber JS. Exposure-response relationships of the efficacy and safety of ipilimumab in patients with advanced melanoma. Clin Cancer Res. (2013) 19:3977–86. doi: 10.1158/1078-0432.Ccr-12-3243

PubMed Abstract | Crossref Full Text | Google Scholar

28. Kvistborg P, Philips D, Kelderman S, Hageman L, Ottensmeier C, Joseph-Pietras D, et al. Anti-Ctla-4 therapy broadens the melanoma-reactive Cd8+ T cell response. Sci Trans Med. (2014) 6:254ra128. doi: 10.1126/scitranslmed.3008918

PubMed Abstract | Crossref Full Text | Google Scholar

29. Meyers JH, Sabatos CA, Chakravarti S, Kuchroo VK. The Tim gene family regulates autoimmune and allergic diseases. Trends Mol Med. (2005) 11:362–9. doi: 10.1016/j.molmed.2005.06.008

PubMed Abstract | Crossref Full Text | Google Scholar

30. Monney L, Sabatos CA, Gaglia JL, Ryu A, Waldner H, Chernova T, et al. Th1-specific cell surface protein Tim-3 regulates macrophage activation and severity of an autoimmune disease. Nature. (2002) 415:536–41. doi: 10.1038/415536a

PubMed Abstract | Crossref Full Text | Google Scholar

31. Jiang X, Yu J, Shi Q, Xiao Y, Wang W, Chen G, et al. Tim-3 promotes intestinal homeostasis in Dss colitis by inhibiting M1 polarization of macrophages. Clin Immunol (Orlando Fla). (2015) 160:328–35. doi: 10.1016/j.clim.2015.07.008

PubMed Abstract | Crossref Full Text | Google Scholar

32. Xu LY, Chen DD, He JY, Lu CC, Liu XG, Le HB, et al. Tim-3 expression by peripheral natural killer cells and natural killer T cells increases in patients with lung cancer–reduction after surgical resection. Asian Pacific J Cancer prevention: APJCP. (2014) 15:9945–8. doi: 10.7314/apjcp.2014.15.22.9945

PubMed Abstract | Crossref Full Text | Google Scholar

33. Datar I, Sanmamed MF, Wang J, Henick BS, Choi J, Badri T, et al. Expression analysis and significance of Pd-1, Lag-3, and Tim-3 in human non-small cell lung cancer using spatially resolved and multiparametric single-cell analysis. Clin Cancer Res. (2019) 25:4663–73. doi: 10.1158/1078-0432.Ccr-18-4142

PubMed Abstract | Crossref Full Text | Google Scholar

34. Curigliano G, Gelderblom H, Mach N, Doi T, Tai D, Forde PM, et al. Phase I/Ib clinical trial of sabatolimab, an anti-Tim-3 antibody, alone and in combination with spartalizumab, an anti-Pd-1 antibody, in advanced solid tumors. Clin Cancer Res. (2021) 27:3620–9. doi: 10.1158/1078-0432.Ccr-20-4746

PubMed Abstract | Crossref Full Text | Google Scholar

35. Roy D, Gilmour C, Patnaik S, Wang LL. Combinatorial blockade for cancer immunotherapy: targeting emerging immune checkpoint receptors. Front Immunol. (2023) 14:1264327. doi: 10.3389/fimmu.2023.1264327

PubMed Abstract | Crossref Full Text | Google Scholar

36. Crise B, Rose JK. Identification of palmitoylation sites on Cd4, the human immunodeficiency virus receptor. J Biol Chem. (1992) 267:13593–7. doi: 10.1016/S0021-9258(18)42253-3

PubMed Abstract | Crossref Full Text | Google Scholar

37. Workman CJ, Rice DS, Dugger KJ, Kurschner C, Vignali DA. Phenotypic analysis of the murine Cd4-related glycoprotein, Cd223 (Lag-3). Eur J Immunol. (2002) 32:2255–63. doi: 10.1002/1521-4141(200208)32:8<2255::Aid-immu2255>3.0.Co;2-a

PubMed Abstract | Crossref Full Text | Google Scholar

38. Triebel F, Jitsukawa S, Baixeras E, Roman-Roman S, Genevee C, Viegas-Pequignot E, et al. Lag-3, a novel lymphocyte activation gene closely related to Cd4. J Exp Med. (1990) 171:1393–405. doi: 10.1084/jem.171.5.1393

PubMed Abstract | Crossref Full Text | Google Scholar

39. Workman CJ, Cauley LS, Kim I-J, Blackman MA, Woodland DL, Vignali DA. Lymphocyte activation gene-3 (Cd223) regulates the size of the expanding T cell population following antigen activation in vivo. J Immunol. (2004) 172:5450–5. doi: 10.4049/jimmunol.172.9.5450

PubMed Abstract | Crossref Full Text | Google Scholar

40. Uboha NV, Milhem MM, Kovacs C, Amin A, Magley A, Purkayastha DD, et al. Phase ii study of spartalizumab (Pdr001) and lag525 in advanced solid tumors and hematologic Malignancies. Am Soc Clin Oncol. (2019) 37(15_suppl):2553-. doi: 10.1200/JCO.2019.37.15_suppl.2553

Crossref Full Text | Google Scholar

41. Jacob S, Daud A. Phase ib/ii study of xmab23104 (Pd1 X icos) and xmab22841 (Ctla-4 X lag3) combination in metastatic melanoma refractory to prior immune checkpoint inhibitor therapy with and without cns disease. J Clin Oncol. (2023) 41:TPS9595–TPS. doi: 10.1200/JCO.2023.41.16_suppl.TPS9595

Crossref Full Text | Google Scholar

42. Stanietsky N, Simic H, Arapovic J, Toporik A, Levy O, Novik A, et al. The interaction of tigit with pvr and pvrl2 inhibits human Nk cell cytotoxicity. Proc Natl Acad Sci U S A. (2009) 106:17858–63. doi: 10.1073/pnas.0903474106

PubMed Abstract | Crossref Full Text | Google Scholar

43. Li M, Xia P, Du Y, Liu S, Huang G, Chen J, et al. T-cell immunoglobulin and itim domain (Tigit) receptor/poliovirus receptor (Pvr) ligand engagement suppresses interferon-Γ Production of natural killer cells via B-arrestin 2-mediated negative signaling*. J Biol Chem. (2014) 289:17647–57. doi: 10.1074/jbc.M114.572420

PubMed Abstract | Crossref Full Text | Google Scholar

44. Johnston Robert J, Comps-Agrar L, Hackney J, Yu X, Huseni M, Yang Y, et al. The immunoreceptor tigit regulates antitumor and antiviral Cd8+ T cell effector function. Cancer Cell. (2014) 26:923–37. doi: 10.1016/j.ccell.2014.10.018

PubMed Abstract | Crossref Full Text | Google Scholar

45. Yang Z, Peng Y, Xu J, Chen P, Zhao Z, Cai Q, et al. Pvr/Tigit and Pd-L1/Pd-1 expression predicts survival and enlightens combined immunotherapy in lung squamous cell carcinoma. Trans Oncol. (2022) 24:101501. doi: 10.1016/j.tranon.2022.101501

PubMed Abstract | Crossref Full Text | Google Scholar

46. Xiao K, Xiao K, Li K, Xue P, Zhu S. Prognostic role of Tigit expression in patients with solid tumors: A meta-analysis. J Immunol Res. (2021) 2021:5440572. doi: 10.1155/2021/5440572

PubMed Abstract | Crossref Full Text | Google Scholar

47. Xu Y, Cui G, Jiang Z, Li N, Zhang X. Survival analysis with regard to Pd-L1 and Cd155 expression in human small cell lung cancer and a comparison with associated receptors. Oncol Lett. (2019) 17:2960–8. doi: 10.3892/ol.2019.9910

PubMed Abstract | Crossref Full Text | Google Scholar

48. Sun Y, Luo J, Chen Y, Cui J, Lei Y, Cui Y, et al. Combined evaluation of the expression status of Cd155 and Tigit plays an important role in the prognosis of luad (Lung adenocarcinoma). Int Immunopharmacol. (2020) 80:106198. doi: 10.1016/j.intimp.2020.106198

PubMed Abstract | Crossref Full Text | Google Scholar

49. Ostroumov D, Duong S, Wingerath J, Woller N, Manns MP, Timrott K, et al. Transcriptome profiling identifies Tigit as a marker of T-cell exhaustion in liver cancer. Hepatology. (2021) 73:1399–418. doi: 10.1002/hep.31466

PubMed Abstract | Crossref Full Text | Google Scholar

50. Niu J, Maurice-Dror C, Lee DH, Kim DW, Nagrial A, Voskoboynik M, et al. First-in-human phase 1 study of the anti-tigit antibody vibostolimab as monotherapy or with pembrolizumab for advanced solid tumors, including non-small-cell lung cancer(☆). Ann Oncol. (2022) 33:169–80. doi: 10.1016/j.annonc.2021.11.002

PubMed Abstract | Crossref Full Text | Google Scholar

51. Watanabe N, Gavrieli M, Sedy JR, Yang J, Fallarino F, Loftin SK, et al. Btla is a lymphocyte inhibitory receptor with similarities to Ctla-4 and Pd-1. Nat Immunol. (2003) 4:670–9. doi: 10.1038/ni944

PubMed Abstract | Crossref Full Text | Google Scholar

52. Gaikwad S, Agrawal MY, Kaushik I, Ramachandran S, Srivastava SK. Immune checkpoint proteins: signaling mechanisms and molecular interactions in cancer immunotherapy. Semin Cancer Biol. (2022) 86:137–50. doi: 10.1016/j.semcancer.2022.03.014

PubMed Abstract | Crossref Full Text | Google Scholar

53. Sedy JR, Gavrieli M, Potter KG, Hurchla MA, Lindsley RC, Hildner K, et al. B and T lymphocyte attenuator regulates T cell activation through interaction with herpesvirus entry mediator. Nat Immunol. (2005) 6:90–8. doi: 10.1038/ni1144

PubMed Abstract | Crossref Full Text | Google Scholar

54. Shui JW, Steinberg MW, Kronenberg M. Regulation of inflammation, autoimmunity, and infection immunity by Hvem-Btla signaling. J leukocyte Biol. (2011) 89:517–23. doi: 10.1189/jlb.0910528

PubMed Abstract | Crossref Full Text | Google Scholar

55. Wang XF, Chen YJ, Wang Q, Ge Y, Dai Q, Yang KF, et al. Distinct expression and inhibitory function of B and T lymphocyte attenuator on human T cells. Tissue Antigens. (2007) 69:145–53. doi: 10.1111/j.1399-0039.2006.00710.x

PubMed Abstract | Crossref Full Text | Google Scholar

56. Ren S, Tian Q, Amar N, Yu H, Rivard CJ, Caldwell C, et al. The immune checkpoint, hvem may contribute to immune escape in non-small cell lung cancer lacking Pd-L1 expression. Lung Cancer (Amsterdam Netherlands). (2018) 125:115–20. doi: 10.1016/j.lungcan.2018.09.004

PubMed Abstract | Crossref Full Text | Google Scholar

57. Mittal R, Chen CW, Lyons JD, Margoles LM, Liang Z, Coopersmith CM, et al. Murine lung cancer induces generalized T-cell exhaustion. J Surg Res. (2015) 195:541–9. doi: 10.1016/j.jss.2015.02.004

PubMed Abstract | Crossref Full Text | Google Scholar

58. Thommen DS, Schreiner J, Müller P, Herzig P, Roller A, Belousov A, et al. Progression of lung cancer is associated with increased dysfunction of T cells defined by coexpression of multiple inhibitory receptors. Cancer Immunol Res. (2015) 3:1344–55. doi: 10.1158/2326-6066.Cir-15-0097

PubMed Abstract | Crossref Full Text | Google Scholar

59. Li X, Xu Z, Cui G, Yu L, Zhang X. Btla expression in stage I-iii non-small-cell lung cancer and its correlation with Pd-1/Pd-L1 and clinical outcomes. OncoTargets Ther. (2020) 13:215–24. doi: 10.2147/ott.S232234

PubMed Abstract | Crossref Full Text | Google Scholar

60. Brown ZJ, Yu SJ, Heinrich B, Ma C, Fu Q, Sandhu M, et al. Indoleamine 2, 3-dioxygenase provides adaptive resistance to immune checkpoint inhibitors in hepatocellular carcinoma. Cancer Immunology Immunotherapy. (2018) 67:1305–15. doi: 10.1007/s00262-018-2190-4

PubMed Abstract | Crossref Full Text | Google Scholar

61. Lim J-Y, Lee S-E, Park G, Choi EY, Min C-K. Inhibition of indoleamine 2, 3-dioxygenase by stereoisomers of 1-methyl tryptophan in an experimental graft-versus-tumor model. Exp Hematol. (2014) 42:862–6.e3. doi: 10.1016/j.exphem.2014.06.006

PubMed Abstract | Crossref Full Text | Google Scholar

62. Asghar K, Farooq A, Zulfiqar B, Rashid MU. Indoleamine 2,3-dioxygenase: as a potential prognostic marker and immunotherapeutic target for hepatocellular carcinoma. World J Gastroenterol. (2017) 23:2286–93. doi: 10.3748/wjg.v23.i13.2286

PubMed Abstract | Crossref Full Text | Google Scholar

63. Schalper KA, Carvajal-Hausdorf D, McLaughlin J, Altan M, Velcheti V, Gaule P, et al. Differential expression and significance of Pd-L1, Ido-1, and B7-H4 in human lung cancer. Clin Cancer Res. (2017) 23:370–8. doi: 10.1158/1078-0432.Ccr-16-0150

PubMed Abstract | Crossref Full Text | Google Scholar

64. Shang K, Wang Z, Hu Y, Huang Y, Yuan K, Yu Y. Gene silencing of indoleamine 2,3-dioxygenase 1 inhibits lung cancer growth by suppressing T-cell exhaustion. Oncol Lett. (2020) 19:3827–38. doi: 10.3892/ol.2020.11477

PubMed Abstract | Crossref Full Text | Google Scholar

65. Yang Y, Liu F, Liu W, Ma M, Gao J, Lu Y, et al. Analysis of single-cell rnaseq identifies transitional states of T cells associated with hepatocellular carcinoma. Clin Trans Med. (2020) 10:e133. doi: 10.1002/ctm2.133

PubMed Abstract | Crossref Full Text | Google Scholar

66. Wilson EB, Brooks DG. The role of Il-10 in regulating immunity to persistent viral infections. Curr Top Microbiol Immunol. (2011) 350:39–65. doi: 10.1007/82_2010_96

PubMed Abstract | Crossref Full Text | Google Scholar

67. Mumm JB, Emmerich J, Zhang X, Chan I, Wu L, Mauze S, et al. Il-10 elicits Ifnγ-dependent tumor immune surveillance. Cancer Cell. (2011) 20:781–96. doi: 10.1016/j.ccr.2011.11.003

PubMed Abstract | Crossref Full Text | Google Scholar

68. Neven B, Mamessier E, Bruneau J, Kaltenbach S, Kotlarz D, Suarez F, et al. A Mendelian predisposition to B-cell lymphoma caused by Il-10r deficiency. Blood. (2013) 122:3713–22. doi: 10.1182/blood-2013-06-508267

PubMed Abstract | Crossref Full Text | Google Scholar

69. Ruffell B, Chang-Strachan D, Chan V, Rosenbusch A, Ho CM, Pryer N, et al. Macrophage Il-10 blocks Cd8+ T cell-dependent responses to chemotherapy by suppressing Il-12 expression in intratumoral dendritic cells. Cancer Cell. (2014) 26:623–37. doi: 10.1016/j.ccell.2014.09.006

PubMed Abstract | Crossref Full Text | Google Scholar

70. Sawant DV, Yano H, Chikina M, Zhang Q, Liao M, Liu C, et al. Adaptive plasticity of Il-10(+) and Il-35(+) T(Reg) cells cooperatively promotes tumor T cell exhaustion. Nat Immunol. (2019) 20:724–35. doi: 10.1038/s41590-019-0346-9

PubMed Abstract | Crossref Full Text | Google Scholar

71. Xu Y, Zeng H, Jin K, Liu Z, Zhu Y, Xu L, et al. Immunosuppressive tumor-associated macrophages expressing interlukin-10 conferred poor prognosis and therapeutic vulnerability in patients with muscle-invasive bladder cancer. J immunotherapy Cancer. (2022) 10(3). doi: 10.1136/jitc-2021-003416

PubMed Abstract | Crossref Full Text | Google Scholar

72. Shiri AM, Zhang T, Bedke T, Zazara DE, Zhao L, Lücke J, et al. Il-10 dampens antitumor immunity and promotes liver metastasis via Pd-L1 induction. J Hepatol. (2024) 80:634–44. doi: 10.1016/j.jhep.2023.12.015

PubMed Abstract | Crossref Full Text | Google Scholar

73. Li L, Yu R, Cai T, Chen Z, Lan M, Zou T, et al. Effects of immune cells and cytokines on inflammation and immunosuppression in the tumor microenvironment. Int Immunopharmacol. (2020) 88:106939. doi: 10.1016/j.intimp.2020.106939

PubMed Abstract | Crossref Full Text | Google Scholar

74. Huang Y, Zou K, Jiang H, Li Z. The complex role of Il-10 in Malignant ascites: A review. Cancer immunology immunotherapy: CII. (2024) 73:32. doi: 10.1007/s00262-023-03616-y

PubMed Abstract | Crossref Full Text | Google Scholar

75. Bengsch B, Johnson AL, Kurachi M, Odorizzi PM, Pauken KE, Attanasio J, et al. Bioenergetic insufficiencies due to metabolic alterations regulated by the inhibitory receptor Pd-1 are an early driver of Cd8+ T cell exhaustion. Immunity. (2016) 45:358–73.

PubMed Abstract | Google Scholar

76. Guo Y, Xie Y-Q, Gao M, Zhao Y, Franco F, Wenes M, et al. Metabolic reprogramming of terminally exhausted Cd8+ T cells by Il-10 enhances anti-tumor immunity. Nat Immunol. (2021) 22:746–56. doi: 10.1038/s41590-021-00940-2

PubMed Abstract | Crossref Full Text | Google Scholar

77. Commins S, Steinke JW, Borish L. The extended Il-10 superfamily: Il-10, Il-19, Il-20, Il-22, Il-24, Il-26, Il-28, and Il-29. J Allergy Clin Immunol. (2008) 121:1108–11. doi: 10.1016/j.jaci.2008.02.026

PubMed Abstract | Crossref Full Text | Google Scholar

78. Banchereau J, Brière F, Liu YJ, Rousset F. Molecular control of B lymphocyte growth and differentiation. Stem Cells (Dayton Ohio). (1994) 12:278–88. doi: 10.1002/stem.5530120304

PubMed Abstract | Crossref Full Text | Google Scholar

79. Shi W, Yao X, Fu Y, Wang Y. Interferon-A and its effects on cancer cell apoptosis. Oncol Lett. (2022) 24:235. doi: 10.3892/ol.2022.13355

PubMed Abstract | Crossref Full Text | Google Scholar

80. Fujimura T, Hidaka T, Kambayashi Y, Furudate S, Kakizaki A, Tono H, et al. Phase I study of nivolumab combined with Ifn-B for patients with advanced melanoma. Oncotarget. (2017) 8:71181–7. doi: 10.18632/oncotarget.17090

PubMed Abstract | Crossref Full Text | Google Scholar

81. McNab F, Mayer-Barber K, Sher A, Wack A, O'Garra A. Type I interferons in infectious disease. Nat Rev Immunol. (2015) 15:87–103. doi: 10.1038/nri3787

PubMed Abstract | Crossref Full Text | Google Scholar

82. Marshall HD, Urban SL, Welsh RM. Virus-induced transient immune suppression and the inhibition of T cell proliferation by type I interferon. J Virol. (2011) 85:5929–39. doi: 10.1128/jvi.02516-10

PubMed Abstract | Crossref Full Text | Google Scholar

83. Wu T, Ji Y, Moseman EA, Xu HC, Manglani M, Kirby M, et al. The tcf1-bcl6 axis counteracts type I interferon to repress exhaustion and maintain T cell stemness. Sci Immunol. (2016) 1(6). doi: 10.1126/sciimmunol.aai8593

PubMed Abstract | Crossref Full Text | Google Scholar

84. Vardhana SA, Hwee MA, Berisa M, Wells DK, Yost KE, King B, et al. Impaired mitochondrial oxidative phosphorylation limits the self-renewal of T cells exposed to persistent antigen. Nat Immunol. (2020) 21:1022–33. doi: 10.1038/s41590-020-0725-2

PubMed Abstract | Crossref Full Text | Google Scholar

85. Park BV, Freeman ZT, Ghasemzadeh A, Chattergoon MA, Rutebemberwa A, Steigner J, et al. Tgfβ1-mediated Smad3 enhances Pd-1 expression on antigen-specific T cells in cancer. Cancer Discovery. (2016) 6:1366–81. doi: 10.1158/2159-8290.CD-15-1347

PubMed Abstract | Crossref Full Text | Google Scholar

86. Hashimoto M, Araki K, Cardenas MA, Li P, Jadhav RR, Kissick HT, et al. Pd-1 combination therapy with Il-2 modifies Cd8(+) T cell exhaustion program. Nature. (2022) 610:173–81. doi: 10.1038/s41586-022-05257-0

PubMed Abstract | Crossref Full Text | Google Scholar

87. Liu Y, Zhou N, Zhou L, Wang J, Zhou Y, Zhang T, et al. Il-2 regulates tumor-reactive Cd8+ T cell exhaustion by activating the Aryl hydrocarbon receptor. Nat Immunol. (2021) 22:358–69. doi: 10.1038/s41590-020-00850-9

PubMed Abstract | Crossref Full Text | Google Scholar

88. Zhu W, Li Y, Han M, Jiang J. Regulatory mechanisms and reversal of Cd8+ T cell exhaustion: A literature review. Biology. (2023) 12:541. doi: 10.3390/biology12040541

PubMed Abstract | Crossref Full Text | Google Scholar

89. Dai E, Zhu Z, Wahed S, Qu Z, Storkus WJ, Guo ZS. Epigenetic modulation of antitumor immunity for improved cancer immunotherapy. Mol Cancer. (2021) 20:1–27. doi: 10.1186/s12943-021-01464-x

PubMed Abstract | Crossref Full Text | Google Scholar

90. Saucillo DC, Gerriets VA, Sheng J, Rathmell JC, Maciver NJ. Leptin metabolically licenses T cells for activation to link nutrition and immunity. J Immunol (Baltimore Md: 1950). (2014) 192:136–44. doi: 10.4049/jimmunol.1301158

PubMed Abstract | Crossref Full Text | Google Scholar

91. Michalek RD, Rathmell JC. The metabolic life and times of a T-cell. Immunol Rev. (2010) 236:190–202. doi: 10.1111/j.1600-065X.2010.00911.x

PubMed Abstract | Crossref Full Text | Google Scholar

92. Harris SG, Padilla J, Koumas L, Ray D, Phipps RP. Prostaglandins as modulators of immunity. Trends Immunol. (2002) 23:144–50. doi: 10.1016/s1471-4906(01)02154-8

PubMed Abstract | Crossref Full Text | Google Scholar

93. Zelenay S, van der Veen AG, Böttcher JP, Snelgrove KJ, Rogers N, Acton SE, et al. Cyclooxygenase-dependent tumor growth through evasion of immunity. Cell. (2015) 162:1257–70. doi: 10.1016/j.cell.2015.08.015

PubMed Abstract | Crossref Full Text | Google Scholar

94. Cheng Y, Wang M, Yu Y, Lawson J, Funk CD, Fitzgerald GA. Cyclooxygenases, microsomal prostaglandin E synthase-1, and cardiovascular function. J Clin Invest. (2006) 116:1391–9. doi: 10.1172/jci27540

PubMed Abstract | Crossref Full Text | Google Scholar

95. Wang J, Zhang L, Kang D, Yang D, Tang Y. Activation of Pge2/Ep2 and Pge2/Ep4 signaling pathways positively regulate the level of Pd-1 in infiltrating Cd8(+) T cells in patients with lung cancer. Oncol Lett. (2018) 15:552–8. doi: 10.3892/ol.2017.7279

PubMed Abstract | Crossref Full Text | Google Scholar

96. Clayton A, Al-Taei S, Webber J, Mason MD, Tabi Z. Cancer exosomes express Cd39 and Cd73, which suppress T cells through adenosine production. J Immunol (Baltimore Md: 1950). (2011) 187:676–83. doi: 10.4049/jimmunol.1003884

PubMed Abstract | Crossref Full Text | Google Scholar

97. Kjaergaard J, Hatfield S, Jones G, Ohta A, Sitkovsky M. A(2a) adenosine receptor gene deletion or synthetic a(2a) antagonist liberate tumor-reactive Cd8(+) T cells from tumor-induced immunosuppression. J Immunol (Baltimore Md: 1950). (2018) 201:782–91. doi: 10.4049/jimmunol.1700850

PubMed Abstract | Crossref Full Text | Google Scholar

98. Mastelic-Gavillet B, Navarro Rodrigo B, Décombaz L, Wang H, Ercolano G, Ahmed R, et al. Adenosine mediates functional and metabolic suppression of peripheral and tumor-infiltrating Cd8(+) T cells. J immunotherapy Cancer. (2019) 7:257. doi: 10.1186/s40425-019-0719-5

PubMed Abstract | Crossref Full Text | Google Scholar

99. Vijayan D, Young A, Teng MWL, Smyth MJ. Targeting immunosuppressive adenosine in cancer. Nat Rev Cancer. (2017) 17:709–24. doi: 10.1038/nrc.2017.86

PubMed Abstract | Crossref Full Text | Google Scholar

100. Corbet C, Feron O. Tumour acidosis: from the passenger to the driver's seat. Nat Rev Cancer. (2017) 17:577–93. doi: 10.1038/nrc.2017.77

PubMed Abstract | Crossref Full Text | Google Scholar

101. Colegio OR, Chu NQ, Szabo AL, Chu T, Rhebergen AM, Jairam V, et al. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature. (2014) 513:559–63. doi: 10.1038/nature13490

PubMed Abstract | Crossref Full Text | Google Scholar

102. Maj T, Wang W, Crespo J, Zhang H, Wang W, Wei S, et al. Oxidative stress controls regulatory T cell apoptosis and suppressor activity and Pd-L1-blockade resistance in tumor. Nat Immunol. (2017) 18:1332–41. doi: 10.1038/ni.3868

PubMed Abstract | Crossref Full Text | Google Scholar

103. Ikonen E. Cellular cholesterol trafficking and compartmentalization. Nat Rev Mol Cell Biol. (2008) 9:125–38. doi: 10.1038/nrm2336

PubMed Abstract | Crossref Full Text | Google Scholar

104. Ma X, Bi E, Lu Y, Su P, Huang C, Liu L, et al. Cholesterol induces Cd8(+) T cell exhaustion in the tumor microenvironment. Cell Metab. (2019) 30:143–56.e5. doi: 10.1016/j.cmet.2019.04.002

PubMed Abstract | Crossref Full Text | Google Scholar

105. Ma X, Bi E, Huang C, Lu Y, Xue G, Guo X, et al. Cholesterol negatively regulates Il-9-producing Cd8(+) T cell differentiation and antitumor activity. J Exp Med. (2018) 215:1555–69. doi: 10.1084/jem.20171576

PubMed Abstract | Crossref Full Text | Google Scholar

106. Law M, Rudnicka AR. Statin safety: A systematic review. Am J Cardiol. (2006) 97:52c–60c. doi: 10.1016/j.amjcard.2005.12.010

PubMed Abstract | Crossref Full Text | Google Scholar

107. Nielsen SF, Nordestgaard BG, Bojesen SE. Statin use and reduced cancer-related mortality. New Engl J Med. (2012) 367:1792–802. doi: 10.1056/NEJMoa1201735

PubMed Abstract | Crossref Full Text | Google Scholar

108. Takamori S, Toyokawa G, Takada K, Shoji F, Okamoto T, Maehara Y. Combination therapy of radiotherapy and anti-Pd-1/Pd-L1 treatment in non-small-cell lung cancer: A mini-review. Clin Lung Cancer. (2018) 19:12–6. doi: 10.1016/j.cllc.2017.06.015

PubMed Abstract | Crossref Full Text | Google Scholar

109. McLane LM, Abdel-Hakeem MS, Wherry EJ. Cd8 T cell exhaustion during chronic viral infection and cancer. Annu Rev Immunol. (2019) 37:457–95. doi: 10.1146/annurev-immunol-041015-055318

PubMed Abstract | Crossref Full Text | Google Scholar

110. Zhou G, Sprengers D, Boor PPC, Doukas M, Schutz H, Mancham S, et al. Antibodies against Immune Checkpoint Molecules Restore functions of Tumor-Infiltrating T Cells in Hepatocellular carcinomas. Gastroenterology. (2017) 153:1107–19.e10. doi: 10.1053/j.gastro.2017.06.017

PubMed Abstract | Crossref Full Text | Google Scholar

111. Wright K. Fda approves nivolumab plus ipilimumab for the treatment of advanced Hcc. Oncol (Williston Park NY). (2020) 34:693606–.

PubMed Abstract | Google Scholar

Keywords: T cell exhaustion, immunotherapy, non-small cell lung cancer, cytokines, immune checkpoint inhibitors

Citation: Liu X, Xi X, Xu S, Chu H, Hu P, Li D, Zhang B, Liu H, Jiang T and Lu Z (2024) Targeting T cell exhaustion: emerging strategies in non-small cell lung cancer. Front. Immunol. 15:1507501. doi: 10.3389/fimmu.2024.1507501

Received: 07 October 2024; Accepted: 22 November 2024;
Published: 12 December 2024.

Edited by:

Xiangyu Chen, Chongqing Medical University, China

Reviewed by:

Sheefa Mirza, University of the Witwatersrand, South Africa

Copyright © 2024 Liu, Xi, Xu, Chu, Hu, Li, Zhang, Liu, Jiang and Lu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Shengshan Xu, eHVzaGVuZ3NoYW45N0AxNjMuY29t; Tianxiao Jiang, dGlhbnhpYW8uamlhbmdAbWVkLnVuaS1tdWVuY2hlbi5kZQ==; Zhuming Lu, bHptMjE5QGpudS5lZHUuY24=

These authors have contributed equally to this work and share first authorship

ORCID: Xianqiang Liu, orcid.org/0009-0006-7094-2177

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.