Next Article in Journal
Molecular Mechanisms in the Transformation from Indolent to Aggressive B Cell Malignancies
Previous Article in Journal
Granulocyte-Macrophage Colony Stimulating Factor Receptor Contributes to Plexiform Neurofibroma Initiation
Previous Article in Special Issue
Methylation Biomarkers of Lung Cancer Risk: A Systematic Review and Meta-Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting Immune Checkpoint Inhibitors for Non-Small-Cell Lung Cancer: Beyond PD-1/PD-L1 Monoclonal Antibodies

by
Nicolas Roussot
1,2,3,4,*,
Courèche Kaderbhai
1 and
François Ghiringhelli
1,2,3,4
1
Department of Medical Oncology, Centre Georges-François Leclerc, 21000 Dijon, France
2
Cancer Biology Transfer Platform, Centre Georges-François Leclerc, 21000 Dijon, France
3
Centre de Recherche INSERM LNC-UMR1231, Team TIRECs: Therapies and Immune REsponse in CancerS, 21000 Dijon, France
4
UFR Sciences de Santé, Université de Bourgogne, 21000 Dijon, France
*
Author to whom correspondence should be addressed.
Cancers 2025, 17(5), 906; https://doi.org/10.3390/cancers17050906
Submission received: 31 January 2025 / Revised: 28 February 2025 / Accepted: 3 March 2025 / Published: 6 March 2025

Simple Summary

This review provides the latest data regarding the clinical development of new immune checkpoints inhibitors for the treatment of non-small-cell lung cancer, beyond the PD-1 and PD-L1 targets.

Abstract

Non-small-cell lung cancer (NSCLC) remains a leading cause of cancer-related mortality worldwide. Immunotherapy targeting the PD-1/PD-L1 axis has revolutionized treatment, providing durable responses in a subset of patients. However, with fewer than 50% of patients achieving significant benefits, there is a critical need to expand therapeutic strategies. This review explores emerging targets in immune checkpoint inhibition beyond PD-1/PD-L1, including CTLA-4, TIGIT, LAG-3, TIM-3, NKG2A, and CD39/CD73. We highlight the biological basis of CD8 T cell exhaustion in shaping the antitumor immune response. Novel therapeutic approaches targeting additional inhibitory receptors (IR) are discussed, with a focus on their distinct mechanisms of action and combinatory potential with existing therapies. Despite significant advancements, challenges remain in overcoming resistance mechanisms and optimizing patient selection. This review underscores the importance of dual checkpoint blockade and innovative bispecific antibody engineering to maximize therapeutic outcomes for NSCLC patients.

1. Introduction

Non-small-cell lung cancer (NSCLC) remains a leading cause of cancer-related mortality worldwide [1]. Despite advancements in conventional therapies, metastatic NSCLC (mNSCLC) continues to pose significant challenges [2]. In recent years, immunotherapy has emerged as a transformative approach, revolutionizing the cancer treatment landscape. By harnessing the body’s immune system to combat cancer cells, immunotherapy offers a promising avenue for improving patient outcomes [3]. In patients without oncogene addiction [4], the first-line regimen for metastatic disease was transformed by the use of anti-PD-1/PD-L1 (aPD-1/aPD-L1) antibodies (mAbs) [5,6,7,8,9]. These agents target the programmed cell death protein 1 (PD-1) and its ligand (PD-L1), which are expressed on cancer and immune cells. By blocking the PD-1/PD-L1 pathway, these antibodies help to restore the immune system’s ability to recognize and attack tumor cells [10,11,12,13]. This concept is based on the capacity of cancer to trigger a particular differentiation of CD8 T cells, called the exhaustion program [14,15]. Antitumor T cells, after recognition of their targets, acquire the exhaustion program, marked by the expression of the transcriptional factor TOX1 and the expression of the checkpoint inhibitor PD-1, as well as the expression of multiple additional inhibitory receptors (IRs) [16,17,18,19].
The development of aPD-1/PD-L1 therapies has marked a significant milestone in the treatment of mNSCLC. Based on PD-L1 expression on cancer cells (tumor positive score (TPS)) patients are treated with aPD-1/PD-L1 therapies alone or in combination with chemotherapies [4]. These treatments have demonstrated remarkable efficacy in a subset of patients, leading to durable response and improved survival. However, it is important to note that fewer than 50% of patients benefit from immunotherapy [2]. Factors such as PD-L1 expression [20], tumor mutational burden (TMB) [21,22,23], and immune cell composition [24,25,26,27] can influence treatment response, suggesting that better patient selection could be achieved. In addition, to address the limitations of current immunotherapy approaches, ongoing research is focused on developing complementary therapies. These may include combinations with chemotherapy, targeted therapies, or other immunotherapeutic agents. In this review, we report current clinical research on combinatory treatments that involved dual checkpoint inhibitor blockade.

2. CD8 T Cell Exhaustion

In the context of cancer or chronic infection, and contrary to acute infection or vaccination, CD8 T cells within the tumor are permanently exposed to cancer cells or to pathogens, thus preventing complete antigen clearance. With such antigen persistence, the T cell receptor (TCR) undergoes chronic signaling that mediates a path of differentiation, gradually leading to CD8 T cell exhaustion (reviewed in references [14,15,28,29]). Although heterogeneous, exhausted CD8 T (CD8 Tex) cells are characterized by a progressive loss of effector functions and proliferative potential, impaired responsiveness to homeostatic cytokines, a shift in the metabolic program, altered epigenetic and transcriptional profile, and high expression of IRs. As a consequence of permanent TCR stimulation, activated CD8 T cells longitudinally progress from precursor CD8 Tex (TCF1, EOMES, and TOX expression) to progenitor CD8 Tex (self-renewal ability) to intermediate CD8 Tex (loss of TCF1, sustained TOX expression) and finally to terminal CD8 Tex cells (sustained TOX expression, high IR expression, low cytokine production).
Biologically, the balance between the two transcription factors TCF-1 [30] (encoded by Tcf7) and TOX [31] (encoded by Tox) mediates entrance into and progression of the exhaustion program. In the early phase of exhaustion, progenitor CD8 Tex cells that express TCF-1 conserve proliferative potential. It is now clear that the cellular proliferation increase and the CD8 Tex cell reinvigoration observed after PD-1 blockade result from the effects on this progenitor CD8 Tex subset [32,33,34]. As the exhaustion program progresses, progenitor CD8 Tex cells lose TCF-1 and acquire TOX to become intermediate CD8 Tex, then terminal CD8 Tex cells. TOX might be considered as the tipping point of exhaustion, since its expression is sufficient to promote all key features of CD8 Tex cells [16,18,19], notably the transcriptional regulation and chromatin remodeling that orchestrates the epigenetic landscape of CD8 Tex cells [17]. This epigenetic imprint further limits the anti PD-1 ability to restore effector or memory T cell phenotypes [35,36,37]. Indeed, transcriptomic and epigenetic characterization of CD8 T cells at a single-cell scale clearly distinguished distinct epigenetic features for the canonical CD8 genes Tcf7, Tox, and also Pdcd1 (encoding the IR PD-1) between CD8 T effector, CD8 T memory, and CD8 Tex subsets. To go deeper into the CD8 Tex granularity, chromatin accessibility analyses revealed the existence of distinct epigenetic subsets among the CD8 Tex population. Regarding the impact of immunotherapy within these distinct CD8 Tex subsets, PD-L1 blockade accelerates the differentiation from progenitor CD8 Tex to terminal CD8 Tex cells [38]. Recent studies have deciphered the role of distinct CD8 Tex IRs, demonstrating that IR signaling is not redundant and may synergize to regulate exhaustion features [39,40,41]. Indeed, while PD-1 restrains cellular proliferation, LAG-3 reduces cytokine production, sustains TOX expression, and upregulates the inhibitory CD94/NKG2A receptor through a LAG-3/TOX/CD94/NKG2 axis [42]. Such biological discrepancies between IRs may explain the synergic effect observed with PD-1 and LAG-3 co-blockade [43], notably in NSCLC [44,45]. At the proteome scale, PD-1-positive LAG-3-positive CD8 Tex display a specific enrichment in ubiquitination pathways, especially the E3 ubiquitin ligase Casitas B cell lymphoma (Cbl-b) [46,47].
Pan-cancer analysis of T cells across 16 human cancer types uncovered a T cell transcriptional state characterized by a stress response signature. These stressed CD8 T cells are detectable across distinct cancer types and are enriched in tumors non-responsive to anti-PD1 therapy, notably in NSCLC [48]. Recently, ADRB1, the b1 adrenergic receptor, has been described as a new immune checkpoint and exhaustion marker, as it is expressed by terminal CD8 Tex, but not progenitor CD8 Tex cells, and it promotes exhaustion through an ADRB1-cAMP-CREM pathway, thus suggesting that stress-associated catecholamines participate in CD8 T cell exhaustion [49,50]. Apart from the sympathetic-induced stress response, mechanical stress due to a dense stroma in the tumor microenvironment (TME) induces the transcription factor Osr2 and also mediates CD8 T cell exhaustion [51]. These emerging data uncover the existence of unexpected signaling pathways in the exhaustion program. In the past decade, the main strategy developed to reinvigorate CD8 Tex cells has been to engineer mAb blocking IRs. This review further focuses on the targeting of IRs, namely CTLA-4, LAG-3, TIGIT, TIM-3, CD94/NKG2A, and CD39/CD73.

3. CTLA-4 Targeting

Cytotoxic T lymphocyte-associated protein 4 (CTLA-4) is a pivotal immune checkpoint [52] that plays a central role in regulating T cell activation and maintaining immune homeostasis [53]. It is expressed predominantly on activated T cells and regulatory T cells (Tregs) [54]. CTLA-4 competes with the co-stimulatory receptor CD28 for binding to B7 ligands (CD80 and CD86) on antigen-presenting cells (APC), thereby transmitting an inhibitory signal that limits T cell proliferation and cytokine production [55] (Figure 1). By dampening T cell responses, CTLA-4 prevents autoimmunity but also facilitates tumor immune evasion. Inhibiting CTLA-4 has been a groundbreaking strategy in cancer immunotherapy, aiming to unleash T cell-mediated antitumor immunity [54]. The first-in-class anti-CTLA-4 (aCTLA-4) [56] mAb, ipilimumab, demonstrated durable clinical benefits in melanoma, marking a paradigm shift in oncology and earning FDA approval in 2011. Subsequently, other CTLA-4 targeting agents, such as tremelimumab, have been developed and explored for use in diverse malignancies [57].
The mechanisms of action seem to be multifunctional and involve the capacity of CTLA-4 blockade to improve effector T cell function and increase the proliferation and activation of effector CD4+ and CD8+ T cells. This leads to increased cytokine production and cytotoxic activity, promoting tumor cell eradication. CTLA-4 is preferentially expressed on immunosuppressive Tregs [58]. Anti-CTLA-4 therapies may contribute to antitumor efficacy through the selective depletion of intratumoral Tregs in an Fc receptor-dependent manner [59]. Nevertheless, aCTLA-4 mAbs that lack a crystallizable fragment (Fc) still induce tumor regression, thus suggesting Fc-independent functions [60,61]. CTLA-4 inhibition expands the repertoire of T cells capable of recognizing diverse tumor antigens [62]. This increase in clonal diversity is critical for robust and sustained antitumor responses. Finally, by blocking CTLA-4, these therapies restore the CD28-mediated co-stimulatory signals required for optimal T cell activation and survival. Published results from clinical trials with aCTLA-4 therapies are summarized in Table 1.

3.1. Ipilimumab

Ipilimumab is a fully human IgG1 monoclonal antibody and was the first CTLA-4 inhibitor to demonstrate significant survival benefits in advanced melanoma. Early clinical trials reported that ipilimumab improved overall survival (OS) by several months compared to standard treatments, with a subset of patients achieving long-term remission. Ipilimumab induces durable responses, with some patients achieving survival beyond 10 years [56]. In addition to melanoma, ipilimumab has shown efficacy in other cancers, including NSCLC [63], renal cell carcinoma [64], mesothelioma [65], hepatocellular carcinoma [66], and microsatellite instable (MSI-high) colorectal cancer [67], often in combination with other checkpoint inhibitors like aPD-1 agents [68]. While durable responses have been observed with immune checkpoint inhibitor (ICI) monotherapies, such as ipilimumab, only about 20% of patients respond to single-agent ipilimumab in melanoma. This limitation has driven research into combination therapies to enhance efficacy. Clinically, combining checkpoint inhibitors—such as aCTLA-4 and aPD-1/PD-L1—has shown higher response rates compared to monotherapy with either agent across multiple tumor types. Notably, this combination has demonstrated improved OS in melanoma in comparison to aPD-1 alone or ipilimumab alone [69,70]. Additionally, aCTLA-4 and aPD-1 combination therapy has also exhibited antitumor activity in patients who failed aPD-(L)1 monotherapy, suggesting its potential to overcome adaptive resistance mechanisms in select cases. However, since not all patients respond to the combination therapy, identifying novel mechanisms underlying primary and adaptive resistance is essential to guide rational therapeutic strategies. In a first-line setting for mNSCLC, the CheckMate-9LA [71] trial tested nivolumab plus ipilimumab with two cycles of chemotherapy vs. chemotherapy alone. Immunotherapy strongly improved OS (hazard ratio (HR), 0.73; 95% confidence interval (CI), 0.62–0.85; 5-year OS rates, 18% vs. 11%), regardless of tumor PD-L1 expression or histology or presence of baseline brain metastases. The five-year duration of response (DOR) rate was 19% [72]. The CheckMate-227 [63] trial, which tested nivolumab plus ipilimumab without chemotherapy vs. chemotherapy for PD-L1-positive patients, also demonstrated that the combination of aCTLA-4 with aPD-1 improved OS in this population [73]. These data support the long-term efficacy of this combination for mNSCLC.

3.2. Tremelimumab

Tremelimumab is a human IgG2 anti-CTLA-4 mAb, as developed to address the toxicity concerns associated with ipilimumab. Though initial studies in melanoma did not meet primary endpoints [74], tremelimumab has shown promise in combination regimens. Currently, tremelimumab, combined with the aPD-L1 mAb durvalumab, is approved for unresectable hepatocellular carcinoma in a first-line regimen for metastatic disease [75]. In contrast, in NSCLC, the POSEIDON trial failed to demonstrate a significant improvement in OS with the combination of chemotherapy plus durvalumab and tremelimumab [76]. The particularity of the CTLA-4 blockade is the dose effect of the drug. Indeed, aCTLA-4 therapies are associated with significant toxicity, including colitis, dermatitis, and endocrine disorders. These immune-related adverse events (irAEs) result from immune overactivation and are usually dose-dependent [77]. Strategies such as dose reduction (e.g., 1 mg/kg vs. 3 mg/kg for ipilimumab) and intermittent dosing have been explored to mitigate these effects [78]. However, the efficacy of the CTLA-4 blockade used in monotherapy or in combination with aPD-1/aPD-L1 also seems dose-dependent.

3.3. Botensilimab

Botensilimab is a promising Fc-enhanced aCTLA-4 mAb designed to address the limitations of the conventional CTLA-4 blockade, with the aim of improving treatment efficacy in “cold” or refractory tumors. Fc-optimized mAbs are being engineered to enhance Treg depletion, while minimizing systemic toxicity. Fc-optimized antibodies selectively deplete intratumoral Tregs, improving therapeutic efficacy. Botensilimab demonstrates enhanced binding to activating Fcγ receptors (FcγRs) on APC and natural killer (NK) cells, optimizing T cell priming, activation, and memory formation. It also promotes intratumoral regulatory T cell depletion through mechanisms like antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis [79,80]. This treatment showed promising efficacy in phase I studies in immune refractory tumor and in microsatellite stable colorectal cancer known to be resistant to classical immunotherapies [81]. Trials are in progress in other solid tumors with only a few results for NSCLC [80].
Another strategy involves the use of bispecific antibodies (bsAbs) targeting both aCTLA-4 and aPD-(L)1.

3.4. Cadonilimab (AK-104)

Cadonilimab is a humanized bsAb that binds concurrently to PD-1 and CTLA-4 with high affinity on tumor-infiltrating lymphocytes (TILs), thereby enhancing antitumor efficacy and also safety [82]. For mNSCLC, the AK104-202 study [83], a multicenter, open-label clinical trial, enrolled 53 patients and evaluated the safety and efficacy of cadonilimab. The patients were split into three groups. Group A consisted of patients who had failed platinum-based doublet chemotherapy and were immunotherapy naïve; Group B and Group C comprised patients who had failed platinum-based doublet chemotherapy and showed primary or acquired resistance to ICI, respectively. The primary endpoint was not met; mOS was 19.61, 4.93, and 13.16 months in Groups A, B, and C, respectively. Only 10% of the patients in Group A had a response, while the ORR was 0% in Groups B and C. Clinical trials with cadonilimab are currently ongoing, such as the AK104-207 (NCT04647344), the AK104-208 (NCT04646330), and the LungCadX studies [84]. Although this drug seems to be less efficient for patients that have already experienced resistance to immunotherapy, the results of these trials are expected to bring new insights regarding cadonilimab’s usefulness in NSCLC treatment strategies.

3.5. Erfonrilimab (KN-046)

Erfonrilimab is an IgG-like bsAb that specifically binds to PD-L1 and CTLA-4. Erfonrilimab showed significant antitumor efficacy in a melanoma xenograft mouse model and in a colon cancer syngeneic mouse model. After the phase I study with erfonrilimab, involving enrolled patients with advanced solid tumors, demonstrated safety and a promising antitumor efficacy [85], a phase II study was conducted in patients with mNSCLC in the first-line setting. The blinded independent review committee (BIRC)-assessed ORR was 46%, with a median progression-free survival (PFS) of 5.8 months and a median OS of 26.6 months [86].

3.6. Volrustomig (MEDI5752)

Volrustomig is also an aPD-1/aCTLA-4 bsAb with data reported from a phase Ib/II trial in the first-line setting for mNSCLC. At the 1500 mg dose, with chemotherapy, this drug induced an ORR of 50.0% vs. 47.6% in patients assigned to pembrolizumab plus chemotherapy. The median PFS (15.1 vs. 8.9 months) and OS (not reached vs. 16.5 months) also favored the volrustomig arm [87]. At the 750 mg dose, with chemotherapy, the ORRs were 43.7% and 65.0% in non-squamous and squamous mNSCLC, respectively [88]. Volrustomig with chemotherapy is currently being evaluated in the first-line setting of PD-L1< 50% mNSCLC in the randomized phase III eVOLVE-Lung02 study, versus pembrolizumab with chemotherapy [89].
Additional bsAbs are under development, such as lorigerlimab and vudalimab.
Table 1. Published data from clinical trials with anti-CTLA-4 monoclonal antibodies or bispecific antibodies in non-small-cell lung cancer.
Table 1. Published data from clinical trials with anti-CTLA-4 monoclonal antibodies or bispecific antibodies in non-small-cell lung cancer.
aCTLA-4 mAbTypeStudyPhaseSettingNIntervention1st EndpointResults with aCTLA-4Ref
Ipilimumabhuman IgG1κCM-9LAIIIMetastatic, 1st line719Nivo (aPD-1) +ipi + CT
vs. CT
OS in overall population
  • mOS 15.8 mo with nivo + ipi + CT vs. 11.0 mo with CT (HR 0.66; 95% CI, 0.55–0.80),
  • 5 y OS 18% with nivo + ipi + CT vs. 11% with CT
  • mPFS 6.7 mo with nivo + ip + CT vs. 5.3 mo with CT (HR 0.70; 95% CI, 0.60–0.83)
  • Grade ≥ 3 TRAE 47% with nivo + ipi + CT
[71,72]
CM-227IIIMetastatic, 1st line1189Nivo + ipi
vs. nivo
vs. CT
OS in PD-L1 ≥ 1% of nivo + ipi vs. CT
  • mOS 17.1 mo with nivo + ipi vs. 14.9 mo with CT (p = 0.007)
  • mPFS 5.1 mo with nivo + ipi vs. 5.6 mo with CT
  • Grade ≥ 3 TRAE 32.8% with nivo + ipi vs. 19.4% with nivo
[63,64,65,66,67,68,69,70,71,72,73]
Tremelimumabhuman IgG2aPOSEIDONIIIMetastatic, 1st line1013Treme + durva (aPD-L1) + CT
vs. durva + CT
vs. CT
PFS and OS of durva + CT vs. CT
  • mOS 13.3 mo with treme + durva + CT vs. 11.7 mo with CT (HR, 0.86; 95% CI, 0.72 to 1.02; p = 0.758)
  • mPFS 5.5 mo with treme + durva + CT vs. 4.8 mo with CT (HR, 0.74; 95% CI, 0.62 to 0.89; p = 0.0009)
  • Grade ≥ 3 TRAE 51.8% with treme + durva + CT vs. 44.6% with durva + CT
[76]
BotensilimabFc-enhanced human IgG1Chand et al.Ia/bMetastatic, refractory3 with NSCLCBoten +/− balsti (aPD-1)Safety
  • ORR = 67%
  • Grade ≥ 3 TRAE 33% with boten vs. 26% with boten + balsti
[80]
aCTLA-4 bsAbTypeStudyPhaseSettingNIntervention1st EndpointResults with aCTLA-4Ref
Cadonilimab (AK-104)CTLA-4/PD-1AK104-202Ib/IIMetastatic, Co A: 2nd line, ICI-naïve
Co B: 2nd line, primary resistance to CT-ICI
Co C: 2nd line, secondary resistance to CT-ICI
30 (Co A)
7 (Co B)
16 (Co C)
CadonORR
  • ORR 10% in Co A, 0% in Co B and C
  • mOS 19.61 m in Co A, 4.93 m in Co B, 13.16 m in Co C
  • Grade ≥ 3 TRAE 11.3%
[83]
Erfonrilimab (KN-046)CTLA-4/PD-1Zhao et al.IIMetastatic, 1st line87Erfon + CTORR, DoR
  • ORR = 46.0%
  • mDoR = 8.1 mo
  • mPFS 5.8 mo, mOS 26.6 mo
  • Grade ≥ 3 TRAE 66.7%
[86]
Volrustomig (MEDI5752)CTLA-4/PD-1Ahn et al., Spigel et al.Ib/IIMetastatic, 1st line139Volru 1500 mg + CT vs. volru 750 mg + CT vs. pembro (aPD-1) + CTORR
  • ORR 50.0% with volru 1500 mg + CT vs. 47.6% with pembro + CT
  • mPFS 15.1 mo with volru 1500 mg + CT vs. 8.9 mo with pembro + CT
  • mPFS NR with volru 1500 mg + CT vs. 16.5 mo with pembro + CT
  • ORR with volru 750 mg + C: 43.7% in non-squamous, 65.0% in squamous NSCLC
  • No safety data
[87,88]
mAb: monoclonal antibody; N: number of patients included; nivo: nivolumab; ipi: ipilimumab; CT: chemotherapy; OS: overall survival; mOS: median overall survival; mo: months; HR: hazard ratio; CI: confidence interval; mPFS: median progression-free survival; 5 y: 5-year; TRAE: treatment-related adverse events; treme: tremelimumab; durva: durvalumab; PFS: progression-free survival; boten: botensilimab; balsti: balstilimab; ORR: objective response rate; bsAb: bispecific antibody; Co: cohort; cadon: cadonilimab; ICI: immune checkpoint inhibitor; erfon: erfonrilimab; DoR: duration of response; mDoR: median duration of response; pembro: pembrolizumab; volru: volrustomig.

4. TIGIT Targeting

TIGIT (also known as WUCAM, VSTM3, or Vsig9) is an inhibitory molecule expressed on CD4 and CD8 T lymphocytes, Treg lymphocytes, and NK lymphocytes [90,91,92]. The expression of TIGIT by Treg lymphocytes is involved in their immunosuppressive functions. Tregs expressing TIGIT present an activated phenotype and express a high level of immunosuppressive markers, such as FoxP3, CTLA-4, PD-1, TIM-3, and LAG-3 [93]. TIGIT binds primarily to the CD155 receptor, also known as the polio virus receptor (PVR), which is expressed on monocytes, dendritic cells (DCs), fibroblasts, endothelial cells, and tumor cells from many cancers [94]. TIGIT also binds, albeit with less affinity, to CD112, also known as Nectin-2, which is expressed on DCs, monocytes, and numerous tumor cell subtypes. TIGIT ligands (CD155 and CD112) are shared with CD96 and CD226, which cause co-inhibition and co-stimulation signals, respectively. TIGIT competes with CD226 for binding to CD155 and presents higher affinity to CD115 than CD226 (Figure 2).
The immunosuppressive action of TIGIT is caused by several mechanisms. Firstly, TIGIT could inhibit CD8 T lymphocyte proliferation by acting directly on the formation of the TCR complex [95]. Secondly, TIGIT could also inhibit NK cells directly and blunt their production of interferon gamma (IFNγ) [96]. Thirdly, TIGIT could block the homodimerization of CD226, thus inhibiting its lymphocyte co-stimulation signal [97]. Finally, TIGIT is expressed on Treg lymphocytes and could induce the production of IL-10, trigger Treg activation, and suppress the production of IL-12 by DCs [98]. In mouse models, blocking TIGIT individually has little effect on survival and tumor growth, but the combination of aPD-L1 and anti-TIGIT (aTIGIT) has shown complete response with prolonged survival, as well as better efficacy than aPD-(L)1 alone [97]. In humans, CD155 overexpression is associated with resistance to ICI treatment in NSCLC [99,100], and TIGIT expression is significantly overexpressed in many cancers, particularly NSCLC [101]. Importantly, mAb-targeting TIGIT, with the property of co-engaging the crystallizable fragment (Fc) with the FcγR fragment, enable an improvement in response via a reduction in tumor volume, probably because of a reduction in Treg infiltrate and an increase in infiltration of NK immune cells and macrophages within the TME, leading to increased cytokine release [79,102]. Based on such data, mAbs are in development in clinical trials. aTIGIT mAbs can be divided into two categories, namely mAbs with a so-called “silent” Fc (Fc-silent, Fcs) and “activated” mAbs (Fc-enabled or Fc-active, Fce), which could trigger antibody-dependent cytotoxicity (ADCC). Fce antibodies are able to interact with several receptors, whereas Fcs antibodies bind to none. The impact of the use of these two antibodies in combination with an aPD-1 agent on the TME and peripheral circulation has recently been demonstrated in murine models and in vitro in humans. Fcs antibodies induce reactivation of tumor-specific CD8 T lymphocytes that had undergone exhaustion, without inducing intratumor and peripheral Treg depletion. In contrast, the Fce antibody induces intratumor and peripheral Treg depletion [103]. Among the aTIGIT antibodies developed or in development, domvanalimab is currently one of the only compounds being tested in phase II and III that belongs to the Fcs family. Other aTIGIT mAbs, such as tiragolumab and vibostolimab, belong to the Fce family. Published results from clinical trials with aTIGIT mAb and bsAb are summarized in Table 2.

4.1. Tiragolumab

CITYSCAPE, the first large-scale trial using aTIGIT mAbs, was a phase II study evaluating the combination of tiragolumab (or placebo) with the aPD-L1 agent atezolizumab as a first-line treatment for locally advanced or mNSCLC in PD-L1-positive patients (TPS score ≥ 1). This study also investigated the impact of tumor PD-L1 as a predictive biomarker of response to the combination of tiragolumab and atezolizumab. The study showed a significant benefit in favor of the study combination on the primary endpoints (PFS and ORR) in the overall population, with results strongly influenced by PD-L1 expression, with more significant results in the TPS > 50% population. An improvement in OS was also observed in the PD-L1 50% group [104]. Tissue sample analyses from this study demonstrated that a high baseline level of intratumoral tumor-associated macrophages (TAM) and Treg cells was associated with improved OS only for patients treated with the tiragolumab and atezolizumab combination. Preclinical experiments in mouse models suggest that aTIGIT mAbs activate the myeloid compartment (TAM, monocytes, and DCs), thus reprogramming exhausted CD8 T cells to a memory-like state [105]. The phase III SKYSCRAPER-01 trial has further evaluated the combination of tiragolumab/atezolizumab versus placebo/atezolizumab, in the first-line treatment for mNSCLC with PD-L1 > 50%. Initial communications from the laboratory on the interim analyses show no benefit in terms of PFS or OS [106]. SKYSCRAPER-06 is also a randomized phase III trial that assessed this tiragolumab/atezolizumab combination associated with chemotherapy versus pembrolizumab plus chemotherapy, in the first-line setting for non-squamous mNSCLC. Recently published results showed no improvement of PFS and OS with the addition of the aTIGIT mAb to the chemotherapy and aPD-L1 combination [107].

4.2. Domvanalimab

Domvanalimab is an Fc-silent humanized IgG1 that targets TIGIT. The ARC-7 phase II trial (NCT04262856) is a randomized phase II study evaluating the use of zimberelimab, an aPD-1 mAb in combination with domvanalimab alone or in combination with etrumadenant (an antagonist of adenosine A2a and A2b receptors on immune cells) in treatment-naive patients with PDL1> 50% (TPS) and no targetable oncogenic addiction. The co-primary endpoints were ORR and PFS. With domvanalimab added to the aPD-1, ORR increased from 12% to 18%, and median PFS from 5.4 to 12.0 months [108]. The phase II ARC-10 study (NCT04736173) is currently evaluating the zimberelimab/domvanalimab combination versus pembrolizumab alone in locally advanced NSCLC or mNSCLC, not pretreated, with PD-L1 > 50% and without targetable oncogenic addiction. The primary endpoint is PFS with the first results provided by Arcus biosciences [109,110]. Another randomized phase III trial, STAR-121 (NCT05502237), is evaluating the dual combination of zimberelimab/domvanalimab with a platinum-based doublet versus a pembrolizumab/platinum-based doublet. The target population for this study comprises treatment-naive patients with mNSCLC without a targetable mutation and irrespective of PD-L1 expression. The primary endpoints are PFS and OS [111]. Finally, the phase II, VELOCITY-Lung study (NCT05633667) is evaluating three experimental treatment arms in patients diagnosed with mNSCLC without prior treatment or targetable mutations, irrespective of their PD-L1 status: the anti-TROP-2 conjugate antibody sacituzumab-govitecan combined with the dual immunotherapy zimberelimab/domvanalimab, a second experimental arm combining etrumadenant with the zimberelimab/domvanalimab doublet, and, finally, an etrumadenant/zimberelimab arm. The study was launched in March 2023 and is currently recruiting patients [112].

4.3. Vibostolimab

Vibostolimab is a human IgG1 aTIGIT mAb that was first tested in humans in a phase I trial (NCT02964013) with a pan-tumor arm A and an arm B that included patients with mNSCLC who were treatment-naïve or had been pretreated with one or more lines of treatment, including anti PD-(L)1 immunotherapy [113]. This study led to the phase II KEYVIBE-002 study [114] (NCT04725188) that evaluated the combination of pembrolizumab/vibostolimab with or without docetaxel versus standard second-line docetaxel monotherapy in patients with mNSCLC progressing after a standard first-line platinum-based doublet and aPD-(L)1 immunotherapy, with PFS as a primary endpoint. Median PFS was 5.6 months in the pembrolizumab/vibostolimab and docetaxel arm, 2.7 months in the pembrolizumab/vibostolimab arm, and 3.2 months in the docetaxel arm. Two other studies are ongoing, namely KEYVIBE-003 [115] (NCT04738487), with a pembrolizumab/vibostolimab regimen versus pembrolizumab alone in the first-line treatment for patients diagnosed with mNSCLC with tumor PD-L1 > 1% (TPS), with OS as the primary endpoint, and KEYVIBE-007 [116] (NCT05226598) with a pembrolizumab/vibostolimab regimen combined with a platinum-based doublet versus the standard first-line regimen of pembrolizumab and a platinum-based doublet for systemically naive mNSCLC, regardless of PD-L1 status, and with OS as the primary endpoint.
bsAbs that target TIGIT and other immune checkpoints have been developed in clinical trials with results for NSCLC patients.

4.4. Rilvegostomig (AZD2936)

Rilvegostomig is a bsAb targeting both TIGIT and PD-1 that demonstrated promising early signs of tolerability and efficacy in patients with advanced or metastatic PD-L1-positive NSCLC, following progression on at least one prior ICI (part A, B) according to initial findings from the phase 1/2 ARTEMIDE-01 study (NCT04995523) presented at the 2023 ASCO and 2023 ESMO annual meetings [117,118]. The ORR with rilvegostomig was 4.8% and was comprised entirely of partial responses (PR). The median PFS was 2.1 months, and the 6-month disease control rate (DCR) was 31.3% [118]. Rilvegostomig was well tolerated, with no dose-limiting toxicity observed and no grade 4 or 5 treatment-related adverse events (TRAEs) [117]. The ARTEMIDE-01 study also comprised cohorts of ICI-naïve, previously treated patients with mNSCLC and PD-L1 TPS > 1% (part C, D) [119]. The patients with a TPS of 1–49% had an ORR of 29.0%. The patients with a TPS > 50% were randomized between 750 mg and 1500 mg of rilvegostomig every 3 weeks, achieving ORRs of 61.8% and 36.7%, respectively. The 750 mg dose was selected based on these results, on modeling analysis, and on intratumoral receptor occupancy. In flow cytology analyses, doses of rilvegostomig of 210 mg or greater were found to sustain roughly 90% occupancy of PD-1 and TIGIT receptors on peripheral T cells. The ARTEMIDE-01 study continues to enroll participants with a cohort in the first-line setting of mNSCLC (part E). Additionally, AstraZeneca, the developer of rilvegostomig, has announced plans to initiate a phase 3 study to further explore the agent. Rilvegostomig is also being explored in various combinations for gastric cancer (NCT05702229) and in other lung cancer settings (NCT04612751).
Table 2. Published data from clinical trials with anti-TIGIT monoclonal antibodies or bispecific antibodies in non-small-cell lung cancer.
Table 2. Published data from clinical trials with anti-TIGIT monoclonal antibodies or bispecific antibodies in non-small-cell lung cancer.
aTIGIT mAbTypeStudyPhaseSettingNIntervention1st EndpointResults with aTIGITRef
TiragolumabHuman IgG1κCITYSCAPEIIMetastatic, 1st line, PD-L1 ≥ 1%135Tirago + atezo (aPD-L1)
vs. pcb + atezo
ORR, PFS
  • ORR 31·3% with atezo + tirago vs. 16·2% with atezo + pcb (p = 0·031)
  • mPFS 5·4 mo with atezo + tirago vs. 3·6 mo with atezo + pcb (HR 0·57, p = 0·015)
  • Serious TRAE 21% with tirago + atezo vs. 18% with atezo + pcb
[104]
SKYSCRAPER-01IIIMetastatic, 1st line, PD-L1 > 50%534Tirago + atezo vs. pcb + atezoPFS, OS
  • mOS 22.9 mo with atezo + tirago vs. 16.7 mo with atezo + pcb (HR, 0.81; 95% CI, 0.63–1.03)
  • No safety data
[106]
SKYSCRAPER-06IIIMetastatic, 1st line542Tirago + atezo + CT
vs. pembro + CT
PFS, OS
  • mOS 18.9 mo with tirago + atezo + CT vs. 23.1 mo with pembro + CT (HR, 1.3; 95% CI, 1.0–1.7)
  • mPFS 8.3 mo with tirago + atezo + CT vs. 9.9 mo with pembro + CT (HR, 1.3; 95% CI, 1.0–1.6)
  • Grade ≥ 3 TRAE 61% with tirago + atezo + CT vs. 61% with pembro + CT
[107]
DomvanalimabFc-silent humanized IgG1ARC-7IIMetastatic, 1st line, PD-L1 ≥50% 151Domva + zimbe (aPD-1) + etrumadenant
vs. domva + zimbe
vs. zimbe
ORR, PFS
  • ORR 18% with domva + zimbe vs. 12% with zimbe
  • mPFS 12 mo with domva + zimbe vs. 5.4 mo with zimbe (HR 0.55, 95% CI 0.31–1.0)
  • Grade ≥ 3 TEAE 47% with domva + zimbe vs. 58% with zimbe
[108]
ARC-10IIMetastatic, 1st line, PD-L1 ≥50% 98Domva + zimbe
vs. zimbe
vs. CT
PFS
  • mPFS 11.5 mo with domva + zimbe vs. 6.5 mo with zimbe (HR 0.69, 95% CI 0.40–1.18)
  • mOS NR with domva + zimbe vs. 24.4 mo with zimbe (HR 0.64, 95% CI 0.32–1.25)
  • Grade ≥ 3 TRAE 21.1% with domva + zimbe vs. 15.0% with zimbe
[109,110]
VibostolimabHuman IgG1KEYVIBE-002IIMetastatic, 2nd line225MK-7684A (vibo + pembro [aPD-1]) + docetaxel vsMK-7684A (vibo + pembro)
vs. pcb + docetaxel
PFS
  • mPFS (95% CI) was 5.6 mo with vibo + pembro + docetaxel, 2.7 mo with vibo + pembro vs. 3.2 mo with pcb + docetaxel HR 0.77 (95% CI 0.53–1.13; p = 0.0910) for vibo + pembro + docetaxel vs. pcb + docetaxel; HR 1.40 (0.96–2.02), p = 0.9622 for vibo + pembro vs. pcb + docetaxel
  • TRAE of any grade 96.5% with vibo + pembro + docetaxel vs. 60.3% with vibo + pembro
[114]
aTIGIT bsAbTypeStudyPhaseSettingNIntervention1st EndpointResults with aTIGIT BispecificReference
RilvegostomigTIGIT/PD-1ARTEMIDE-01I/IIMetastatic, PD-L1 ≥ 1%, 2nd line, previous ICI (part A, B)83Rilve 750 or 1500 mgSafety
  • ORR = 4.8%
  • DCR = 31.3%
  • mPFS = 2.1 m
  • Grade ≥ 3 TRAE 8.4%
[117,118]
Metastatic, PD-L1 ≥ 1%, ≥2nd line, ICI-naïve (part C, D)96Rilve 750 or 1500 mgORR
  • ORR PD-L1 1–49% with rilve 750 mg: 29%
  • ORR PD-L1 ≥ 50% with rilve 750 mg: 61.8%
  • ORR PD-L1 ≥ 50% with rilve 1500 mg: 36.7%
  • Grade ≥ 3 TRAE 10.5%
[119]
mAb: monoclonal antibody; N: number of patients included; tirago: tiragolumab; atezo: atezolizumab; pcb: placebo; pembro: pembrolizumab; ORR: objective response rate; PFS: progression-free survival; mPFS: median progression-free survival; mo: months; HR: hazard ratio; OS: overall survival; mOS: median overall survival; CI: confidence interval; TRAE: treatment-related adverse events; domva: domvanalimab; zimbe: zimberelimab; CT: chemotherapy; TEAE: treatment-emergent adverse events; NR: not reached; vibo: vibostolimab; pembro: pembrolizumab; bsAb: bispecific antibody; ICI: immune checkpoint inhibitor; rilve: rilvegostomig; DCR: disease control rate.

5. LAG-3 Targeting

Lymphocyte activation gene-3 (LAG-3, reviewed in references [120,121,122]), also known as CD223, is a type I transmembrane protein located on human activated CD4 T cells, CD8 T cells, NK cells, gamma/delta T cells, and plasmacytoid DCs, as well as on rested/activated Tregs. Although LAG-3 and CD4 have a homologous structure, the two receptors share only 20% of their amino acid sequence. Singularly, LAG-3 is a non-ITIM IR that functions through critical “KIEELE” and “EP” motifs on the cytoplasmic tail [123]. LAG-3 on immune T cell lines or NK cells binds to several ligands, among which class II major histocompatibility complex (MHC-II) is the canonical one. Additionally, in the cancer context, LAG-3 also binds fibrogen-like protein 1 (FGL1) released by cancer cells [124], galectin-3 on tumor and tumor-associated stroma cells [125], and LSECtin on tumor cells [126]. Mechanistically, LAG-3 acts as an IR through several immunosuppressive functions (Figure 3). Firstly, it impairs the functionality of immunocompetent adaptive cells. LAG-3 may constitutionally associate with the TCR-CD3 complex in CD4 and CD8 T cells, independently of the presence of MHC-II, although it is the canonical LAG-3 ligand. After TCR stimulation, LAG-3 and TCR colocalization rapidly increases within the immune synapse, further limiting CD4 and CD8 T cell activation and expansion. Mechanistically, the LAG-3 cytoplasmic tail disrupts LcK-CD4 or LcK-CD8 interaction, thereby inhibiting TCR signaling [123,127]. Beyond the membrane receptor structure, LAG-3 is shed at the cell surface in a soluble form (sLAG-3) by ADAM10 on rested/activated T cells, and ADAM17 on activated T cells only. This sLAG-3 release through metalloproteinase cleavage promotes an antitumor immune response and is required for T cell function [128].
Secondly, it impairs innate immune cell functions such as APC. Conversely, LAG-3-expressing Tregs interact with APC to mediate “reverse” signaling that does not require the LAG-3 cytoplasmic tail. Rather, this LAG-3-mediated DC inhibition relies on cell contact with LAG-3-MHC-II interaction, subsequent ERK activation, and SHP1 recruitment [129]. Thirdly, LAG-3 supports immunosuppressive cell functions. LAG-3 expression is critical for Treg suppressor activity [129,130,131,132]. Mechanistically, LAG-3 mediates PI3K-MTORC-AKT inhibition in Foxp3+ Tregs, thus reducing Myc expression and regulating Treg metabolism that allows normal suppressive function [133]. Especially in NSCLC, a transcriptomic study of lung tumor-infiltrating Tregs obtained from patients revealed divergent Treg clusters, among which highly immunosuppressive “activated Tregs” had markedly increased expression of LAG-3 [134].
As described above, the expression of LAG-3 is a well-known marker of T cell exhaustion. In order to impair LAG-3 immunosuppressive functions, LAG-3-targeted mAbs have been designed. Published results from clinical trials with aLAG-3 mAbs and bsAbs are summarized in Table 3.

5.1. Relatlimab (BMS-986016)

Relatlimab is a human IgG4 mAb; it was the first LAG-3 inhibitor developed [135] and was first approved for the treatment of melanoma. In early-stage NSCLC, the randomized phase II NEOPredict trial compared short-course neoadjuvant nivolumab versus short-course neoadjuvant nivolumab + relatilmab association in resectable stage IB–IIA [45,136] (single-level N2, UICC 8th edition). In the experimental arm, 50% of patients had adenocarcinoma, and 30% had squamous cell lung cancer. Stage I, IIA-B, and IIIA represented 33.3%, 56.7%, and 10% of this arm, respectively. Overall, 73.3% had PD-L1-positive disease. The primary endpoint was met, since all patients underwent surgery within 43 days after treatment initiation, in both arms. Regarding secondary outcomes, a higher ORR was observed with relatilmab addition (27% vs. 10%), but no radiological complete response occurred. Pathological complete response was increased from 13 to 17% with relatilmab, as were the rates of major pathological responses (≤10% viable tumor cells; MPR) (from 27 to 30%), while pathological response rates (≤50% viable tumor cells) increased from 60 to 72%. Having PD-L1-positive disease was associated with better pathological response. There was no statistical difference in 12-month DFS or OS, which were excellent in both arms [45]. Immune cell phenotyping revealed that responders had a significant increase in peripheral blood CD8+ and GZM + CD8+ and lower myeloid (neutrophils, monocytes) and Treg tumor infiltration. Regarding gene expression, nivolumab monotherapy induced LAG-3 gene expression, while the combination arm did not. In mNSCLC, the combination of relatlimab, nivolumab, and platinum-based chemotherapy was evaluated in the first-line setting in the phase II RELATIVITY-104 trial. The use of relatlimab led to an increase in ORR from 43.7% with nivolumab–chemotherapy to 51.3% with the addition of LAG-3 mAbs. This benefit was observed in particular in a subgroup of patients with PD-L1 >= 1 and non-squamous histology disease, with an ORR of 58.0 vs. 39.6% in the arm without relatlimab [137].

5.2. Ieramilimab (LAG-525)

Ieramilimab is a humanized IgG4 mAb that contains the S228 hinge-stabilizing mutation, thus blocking the LAG-3–MHC-II interaction with low nanomolar affinity. After having demonstrated safety as a monotherapy or in combination with an aPD-1 in a phase I study [138], ieramilimab was further tested in a phase II study conducted in advanced solid tumors, in which 42 mNSCLC PD-L1-unselected patients were included [139]. While the ORR was 15% among the pretreated ICI-naïve patients, there were no responders among the patients pretreated with aPD-1/PD-L1. The median PFS was 4.2 and 3.9 months for ICI-naive vs. ICI-pretreated NSCLC patients, respectively. The responding patients had a higher T cell-inflamed signature and LAG-3 expression at baseline than the non-responders.

5.3. Favezelimab (MK-4280)

Favezelimab [140] is a humanized IgG4 LAG-3-targeted mAb that binds the interacting site with MHC class II. The Keynote 495/KeyIMPACT [141,142] trial is a biomarker-directed adaptively randomized multi-cohort phase II study in the first-line setting of mNSCLC. Patients were categorized by the 18 immune-related gene signatures that define the T cell-inflamed gene expression profile (TcellinfGEP) [24,25,26] and TMB [22,23] before randomization. One of the study arms consisted of pembrolizumab + favezelimab 200 mg, which was then changed for favezelimab 800 mg. Patient categorization consisted of a TcellinfGEPlowTMBnon-high (group I), TcellinfGEPlowTMBhigh (group II), TcellinfGEPnon-lowTMBnon-high (group III), and TcellinfGEPnon-lowTMBhigh (group IV). With pembrolizumab + favezelimab 800 mg, the ORRs were 27.3% in group 2, 13.6% in group 3, and 50% in group 4. The corresponding mPFS was 4.2, 3.5, and 13 months, respectively, while the corresponding mOS was 24.5, 11 months, and not reached, respectively. When the ORR was evaluated by TMB status alone in this arm, the response rates ranged from 14% in patients with TMBnon-high tumors to 41% in patients with TMBhigh. When the ORR was evaluated by PD-L1 status, the response rates ranged from 17% in patients with TPS < 1% tumors, 42% in patients with TPS ≥ 1%, 33% in patients with TPS 1–49% tumors, and 55% in patients with TPS ≥ 50% tumors. The ORR by dual TMB and PD-L1 status ranged from 11% in patients with PD-L1 TPS < 1% and TMBnon-high tumors to 64% in patients with PD-L1 TPS ≥ 1% and TMBhigh tumors. However, the combination of pembrolizumab + favezelimab did not pass the response rate prespecified efficacy threshold in any dual biomarker-based subgroup in this study.

5.4. Fianlimab (REGN3767)

Fianlimab is a humanized IgG4 antibody mAb that has been evaluated in the first-line setting of mNSCLC. For patients with PD-L1 >= 50%, a three-arm, randomized, phase II/III study (NCT05785767) is assessing fianlimab high dose + cemiplimab (aPD-1), fianlimab low dose + cemiplimab, and placebo + cemiplimab, with ORR and OS as primary endpoints in the phase II and phase III parts, respectively [143]. For PD-L1-unselected patients, a three-arm, phase II/III study (NCT05800015) is ongoing [144]. The phase III part consists of fianlimab high or low dose (according to the dose determined in the phase II) + cemiplimab + platinum-doublet chemotherapy vs. placebo + cemiplimab + chemotherapy. In early-stage NSCLC (stage II-IIIB), a randomized phase II peri-operative study (NCT06161441) is testing the addition of fianlimab on top of a combination of aPD-1 + platinum-doublet chemotherapy.
Other LAG-3-targeted mAbs are currently under development in early phase trials, but safety and efficacy data in NSCLC patients are not yet available. These include miptenalimab (BI 754111) [145,146,147], a humanized IgG4 anti-LAG-3 mAb (NCT03780725, NCT03964233, NCT03156114, NCT03697304, NCT03433898), TSR 033 (NCT03250832) [148], a humanized IgG4 anti-LAG-3 mAb; Sym022 [149], a Fc-inert human monoclonal antibody targeting LAG-3 (NCT04641871, NCT03311412, NCT03489369), and Tuparstobart (ICAGN02385), a humanized Fc-engineered IgG1κ (NCT03538028).
Beyond mAbs, bsAbs targeting both LAG-3 and PD-(L)1 have been engineered, such as FS118 (LAG-3/PD-L1) [150], tomebstomig RO7247769 (LAG-3/PD-1) [151], and tebotelimab MGD013 (LAG-3/PD-1) [152,153]. All these drugs have been evaluated in phase I basket trials comprising mNSCLC, with a low number of patients and with safety as a primary endpoint. The ORRs ranged from 0% to 17.4% in these studies and probably had less efficacy in the ICI-pretreated mNSCLC patients.
Table 3. Published data from clinical trials with anti-LAG-3 monoclonal antibodies or bispecific antibodies in non-small-cell lung cancer.
Table 3. Published data from clinical trials with anti-LAG-3 monoclonal antibodies or bispecific antibodies in non-small-cell lung cancer.
aLAG-3 mAbTypeStudyPhaseSettingNIntervention1st EndpointResults with aLAG-3Ref
Relatlimab
(BMS-986016)
Human IgG4NEOPredictIIEarly, resectable, stage IB–IIA (single-level N2, UICC 8th edition)60Nivo (aPD-1) + relavs. nivoFeasibility of surgery within 43 days
  • Surgery within 43 days: 100%
  • ORR 27% with nivo + rela % vs. 10% with nivo
  • pCR 17% with nivo + rela % vs. 13% with nivo
  • MPR 30% with nivo + rela % vs. 27% with nivo
  • PR 72% with nivo + rela % vs. 60% with nivo
  • Grade ≥ 3 AE 53% with nivo + rela vs. 40% with nivo
[45]
RELATIVITY-104IIMetastatic, 1st line309Nivo + rela + CT vs. nivo + CTORR
  • ORR 51.3% with nvo + rela + CT vs. 43.7% with nivo + CT
  • mDoR 10.1 mo with nvo + rela + CT vs. 9.1 mo with nivo + CT
  • Grade ≥ 3 TRAE 54% with nivo + rela + CT vs. 55% with nivo + CT
[137]
Ieramilimab
(LAG-525)
Humanized IgG4Lin et al.IIMetastatic, ≥2nd line42 with NSCLCIera + sparta (aPD-1)ORR
  • ORR 15%
  • mPFS 4.2 mo for ICI-naive and 3.9 months for ICI-pretreated patients
  • Serious AE 8.5% in ICI-naïve vs. 2.2% in ICI-pretreated patients
[139]
Favezelimab
(MK-4280)
Humanized IgG4KEYNOTE-495/KeyImPaCTIIMetastatic, 1st line243Pembro (aPD-1) +lenvatinib
vs. pembro + quavon (aCTLA-4)
vs. pembro + fave 800 mg
ORR
  • ORR 29.4%
  • ORR 27.3% in TcellinfGEPlowTMBhigh, 13.6% in TcellinfGEPnon-lowTMBnon-high, 50% in TcellinfGEPnon-lowTMBhigh.
  • mPFS 4.2 mo in TcellinfGEPlowTMBhigh, 3.5 mo in TcellinfGEPnon-lowTMBnon-high, 13 mo in TcellinfGEPnon-lowTMBhigh.
  • mOS 24.5 mo in TcellinfGEPlowTMBhigh, 11 mo in TcellinfGEPnon-lowTMBnon-high, NR in TcellinfGEPnon-lowTMBhigh.
  • Grade ≥ 3 TRAE with 68.8% pembro + lenvatinib vs. 23.2% with pembro + quavon vs. 21.6% with pembro + fave 800 mg
[141,142]
aLAG-3 bsAbTypeStudyPhaseSettingNIntervention1st endpointResults with aLAG-3Ref
FS118LAG-3/PD-L1Yap et al.IMetastatic, ≥2nd line9 with NSCLCFS118Safety
  • ORR = 0%
  • DCR = 33.3%
  • Grade ≥ 3 TRAE 4.7%
[150]
Tebotelimab (MGD013)LAG-3-/PD-1Luke et al.IMetastatic, ≥2nd line35 with NSCLCTeboSafety
  • ORR = 14% in ICI-naïve
  • ORR = 0% in ICI-pretreated
  • Grade ≥ 3 TRAE 22.3%
[152,153]
mAb: monoclonal antibody; N: number of patients included; nivo: nivolumab; rela: relatlimab; ORR: objective response rate; pCR: pathological complete response; MPR: major pathological response; PR: pathological response; AE: adverse events; TRAE: treatment-related adverse events; CT: chemotherapy; mDoR: median duration of response; mo: months; iera: ieramilimab; sparta: spartalizumab; mPFS: median progression-free survival; pembro: pembrolizumab; quavon: quavonlimab; fave: favezelimab; TMB: tumor mutational burden; bsAb: bispecific antibody; DCR: disease control rate; ICI: immune checkpoint inhibitor.

6. TIM-3 Targeting

T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3, reviewed in references [154,155,156]) is a type I transmembrane immunoregulatory protein that belongs to the TIM family. TIM-3 is located on human activated CD4 T cells, CD8 T cells [157], NK cells [158], Tregs [159], myeloid cells [160] (such as DCs, intratumoral monocytes, and macrophages [161]), and also mast cells [162]. The immunoglobulin variable (IgV)-like domain and the mucin domain compose the TIM-3 extracellular domain, which binds cognate ligands. The TIM-3 cytoplasmic tail contains Y256 and Y263 (Y265 and Y272 in humans) residues that interact with HLA-B-associated transcript 3 (BAT3) and the Src family tyrosine kinases LCK and FYN. TIM-3 on immune T cell lines, myeloid cells, and NK cells binds to several ligands: galectin-9 [163,164,165,166] and CEACAM1 on tumor cells/T cell lines/NK cells/myeloid cells (DCs, monocytes, and macrophages) [167,168,169], phosphatidylserine [170] on apoptotic cells, and extracellular high-mobility group box 1 (HMGB1) [171].
TIM-3 acts as an IR through several mechanisms (Figure 4): firstly by impairing functionality of immunocompetent adaptive cells. BAT3 is an adaptor protein located on the TIM-3 cytoplasmic tail that negatively regulates TIM-3 in the absence of binding ligands [172]. Galectin-9 or CEACAM1 binding on TIM-3 T cells induces phosphorylation of Y256 and Y263 residues [167,173], which releases BAT3, thus allowing TIM-3 to exert its inhibitory function. Moreover, the Src-related kinase FYN may participate in TIM-3-mediated suppressive signaling, since it binds the cytoplasmic tail and induces T cell anergy [174,175]. Galectin-9 ligation to TIM-3 can also recruit CD45 and CD148 phosphatases and disrupt the immunological synapse to further inhibit T cell functionality [176]. Secondly, TIM-3 acts by impairing innate immune cell functions, such as APC. HGMB1 mediates danger signaling through ligation on several membrane receptors, such as Toll-like receptor 2 (TLR2), TLR4, and RAGE. In particular, HMGB1 picks up nucleic acid, binds to RAGE, and triggers immunostimulatory functions through the TLR7/9 endosomal pathway [177]. TIM-3 on DCs acts as a molecular sink to sequester HMGB1 and prevents DNA capture by the alarmin [171], thus impairing DNA sensing, notably though the cGAS/STING pathway [178]. TIM-3 on DCs also negatively regulates inflammasome activation, thus decreasing the maintenance of CD8+ effector and stem-like T cells [156]. TIM-3 on DCs links with apoptotic material through phosphatidylserine and may be important for cross-presentation to CD8 T cells, a critical mechanism for ensuring peripheral tolerance [179,180,181]. Thirdly, TIM-3 acts by strengthening immunosuppressive cells. In NSCLC, a majority of intratumoral TIM-3+ CD4+ are Tregs [159,182]. Moreover, TIM-3+ Tregs are highly suppressive and produce immunosuppressive cytokines, such as IL-10 [183,184].
Regarding exhaustion, TIM-3 T cells are considered as terminally differentiated exhausted T cells [185]. Notably, expression of the transcription factor TCF1, which is critical for generation and maintenance of the pool of progenitor CD8 T cells, varies in the opposite manner to TIM-3. Indeed, progenitors of exhausted CD8 T cells are TCF1+ TIM-3-, while the terminally exhausted subset is characterized by high expression of inhibitory molecules (TIM-3+ PD-1) [34,186,187,188]. Published results from clinical trials with aTIM-3 mAbs and bsAbs are summarized in Table 4.

6.1. Cobolimab (TSR-022/GSK4069889)

Cobolimab is a humanized IgG4 TIM-3-targeted mAb. The phase I basket trial AMBER [189,190] included ICI pre-treated mNSCLC patients. In part 2B, the combination of cobolimab + dostarlimab (aPD-1) demonstrated modest efficacy with an ORR of 8.3% and a DCR of 21.4%. The recommended phase 2 dose (RP2D) for cobolimab was 300 mg, which was the dose with the highest ORR (9.8%) [191]. The randomized, three-arm, phase II/III COSTAR study (NCT04655976) is now evaluating dostarlimab ± cobolimab + docetaxel vs docetaxel as the control arm [191].

6.2. Sabatolimab MGB453

Sabatolimab is a humanized IgG4 TIM-3-targeted mAb. In a phase II study, 17 patients with mNSCLC previously treated with an aPD-(L)1 received a combination of MGB453 + spartalizumab (aPD-1) with 41.2% stable disease (SD) [192].

6.3. LY3321367

LY332136 is a humanized IgG1λ, Fc-null TIM-3-targeted mAb, tested as a monotherapy or in combination with the aPD-L1 LY300054 in advanced solid tumors in a phase Ia/b trial. NSCLC patients who had progressive disease as the best immunotherapy response in previous lines had worse outcomes (ORR 0%, DCR 34.8%, mPFS 1.9 months) than those who had disease control or response to prior immunotherapy (ORR 7%, DCR 50%, mPFS 7.3 months) [193]. This molecule is currently under investigation in several trials (NCT03099109, NCT02791334).
Other TIM-3-targeted mAbs are currently under development in early phase trials, but safety and efficacy data in NSCLC patients are not yet available. These include sym-023/S095018, an IgG1, with a variant engineered to remove Fc gammaR binding (NCT03311412, NCT03489343, NCT04641871) BMS-986258, a silent IgG1 (NCT03446040), INCAGN02390 [194], a fully human Fc-engineered-silent IgGk1 (NCT03652077, NCT04370704, NCT05287113), and surzebiclimab/BGB-A425, a humanized IgG1 (NCT03744468, NCT03744468).
In addition, bsAbs targeting both TIM-3 and PD-(L)1 have also been evaluated: sabestomig/AZD7789 (TIM-3/PD-1) [195] and LY-3415244 (TIM-3/PD-L1) [196]. Both bsAbs have been evaluated in early phase trials with mNSCLC, with safety as a primary endpoint. The ORR ranges from 0% to 10.5% in these studies. No results have been communicated for lomvastomig/R07121661 (TIM-3/PD-1) as yet (NCT03708328).
Table 4. Published data from clinical trials with anti-TIM-3 monoclonal or bispecific antibodies in non-small-cell lung cancer.
Table 4. Published data from clinical trials with anti-TIM-3 monoclonal or bispecific antibodies in non-small-cell lung cancer.
aTIM-3 mAbTypeStudyPhaseSettingNIntervention1st EndpointResults with aTIM-3Ref
Cobolimab (TSR-022/GSK4069889)Humanized IgG4AMBERIMetastatic, ≥2nd line after aPD-1/PD-L184Cobo + dostar (aPD-1)ORR
  • ORR 8.3%
  • DCR 21.4%
  • Grade ≥ 3 TRAE 13.1%
[189,190]
Sabatolimab (MGB453)Humanized IgG4Mach et al.IMetastatic, ≥2nd line after aPD-1/PD-L117 with NSCLCSaba + sparta (aPD-1)ORR
  • SD 41.2%
  • Grade ≥ 3 TRAE 11.8%
[192]
LY3321367Humanized IgG1λ, Fc-nullHarding et al.IMetastatic, ≥2nd line 65 with NSCLCLY3321367 +/−LY300054 (aPD-L1)Safety, RP2D
  • PD as the best immunotherapy response in previous lines: ORR 0%, DCR 34.8%, mPFS 1.9 mo
  • SD or PR as the best immunotherapy response in previous lines: ORR 7%, DCR 50%, mPFS 7.3 mo
  • Grade ≥ 3 TRAE 3%with LY3321367 monotherapy
[193]
aTIM-3 bsAbTypeStudyPhaseSettingNIntervention1st EndpointResults with aLAG-3Ref
Sabestomig (AZD7789)TIM-3-/PD-1Besse et al.I/IIaMetastatic, ≥2nd line, ICI-pretreated39SabeSafety
  • ORR = 10.5%
  • DCR = 36.8%
  • Grade ≥ 3 TRAE 0%, TRAE of any grade 41%
[195]
LY3415244TIM-3/PD-L1J1C-MC-JZDAIMetastatic, ≥2nd line2 with NSCLCLY3415244Safety, RP2D
  • ORR = 0%
  • Serious AE 16.7%
[196]
mAb: monoclonal antibody; N: number of patients included; cobo: cobolimab; dostar: dostarlimab; ORR: objective response rate; DCR: disease control rate; TRAE: treatment-related adverse events; saba: sabatolimab; sparta: spartalizumab; SD: stable disease; RP2D: recommended phase 2 dose; PD: progressive disease; mPFS: median progression-free survival; mo: months; PR: partial response; bsAb: bispecific antibody; sabe: sabestomig; AE: adverse events.

7. NKG2A Targeting

NKG2A (reviewed in reference [197]) is a transmembrane immunoregulatory protein that belongs to the NKG2 family (C-type lectins) [198]. NKG2A is expressed on NK, NKT, CD8 T, and γδ T cells, dimerizes with CD94, and has for a cognate ligand the non-classical class I major histocompatibility complex (MHC-I) molecules of the human leukocyte antigen (HLA)-E. The NKG2A cytoplasmic tail contains two immunoreceptor tyrosine-based inhibitory motifs, making this receptor an ITIM receptor [199,200,201].
NKG2A acts as an IR through several mechanisms [202,203,204] (Figure 5). Firstly, by impairing functionality of immunocompetent adaptive cells: on NKG2A+ activated CD8 T cells, the ligation NKG2A-HLA-E axis induces ITIM domain phosphorylation, which recruits the SHP-1 and SHP-2 tyrosine phosphatases [205,206,207,208]. Chronologically, the upregulation of NKG2A by CD8 T cells seems to occur after repeated antigen stimulation. NKG2A may be considered as a later immune checkpoint than PD-1, LAG-3, and TIGIT [209]. Secondly, by impairing innate immune cell functions: as described above, on NKG2A+ NK cells, HLA-E binding protects target cells from lysis [210]. Notably, on NK cells, HLA-E can also bind to the CD94/NKG2C heterodimer. Contrary to NKG2A, NKG2C is an ITAM receptor that cooperates with DAP12 to trigger an activating signal [211]. However, the binding affinity of CD94/NKG2A is approximatively six times higher than CD94/NKG2C [212,213]. The balance between the signaling of these two contradictory receptors may also depend on the type of HLA-E-presented peptide on the target cell [214,215,216]. Thirdly, by modulating the TME: NK cells might influence the polarization of immune cells in the TME through its NKG2 receptors. Indeed, activation of NK cells may trigger a TAM switch by favoring differentiation of a TAM1 subset [217]. Together, these data suggest that targeting the CD94/NKG2A-HLA-E axis may enhance immunotherapy efficacy. Published results from clinical trials with aNKG2A mAbs are summarized in Table 5.

Monalizumab

Monalizumab is a humanized IgG4-blocking mAb specific to NKG2A (it does not bind NKG2C) [218]. In the neoadjuvant setting, among 83 patients enrolled in the phase II NeoCOAST [219] platform trial, 20 received a single cycle of monalizumab with durvalumab before surgery for stage IA3-IIIA NSCLC. With monalizumab added to aPD-L1, the rate of major pathologic response (MPR) increased from 11.1% to 30.0%, while the pCR rate increased from 3.7% to 10%. Interestingly, the density of NKG2A+ cells in the tumor area before treatment was not associated with MPR. RNA sequencing of both pre- and post-treatment specimens showed that NK- and CD8 T cell-associated gene expression was increased in the combination arm. In a more advanced setting, the phase II open-label randomized COAST study [220,221] enrolled unresectable stage III NSCLC patients and assessed the addition of monalizumab or oleclumab (aCD73 mAb) to adjuvant durvalumab after chemoradiation. Monalizumab in addition to durvalumab increased ORR from 23.9% with durvalumab monotherapy to 40.3%, and prolonged mPFS to 19.8 months, versus 7.3 months with durvalumab monotherapy. The combination might also prolong OS (data not mature). On the basis of these results, the combination of monalizumab or oleclumab with durvalumab is currently being evaluated in the double-blind, randomized, placebo-controlled PACIFIC-9 study [222]. In the metastatic setting, the monalizumab and durvalumab combination was assessed in a phase I/II basket study with 20 previously treated but immunotherapy-naïve mNSCLC patients. In this population, aNKG2A with durvalumab showed modest efficacy, with ORR, mPFS, and mOS reaching 10%, 1.9 months, and 8.8 months, respectively. Regarding immune activation in the periphery, the addition of monalizumab increased blood levels of CXCR3-related chemokines (CXCL10 and CXCL11). In the tumor, although there was an increased number of proliferative T (CD3+ Ki67+) and cytotoxic cells (GZMB+) with the combination, no significant difference was observed in NK (Nkp46+) cells [223]. The phase Ib/IIA PIONeeR study [224] tested similar durvalumab and monalizumab therapy, but in mNSCLC patients who had already experienced disease progression after sequential or concomitant PD-(L)1 and platinum-based chemotherapy. The 12-week DCR reached with the combination was 24.1% vs. 54.5% with docetaxel, thus suggesting lower efficacy in this context. Many studies of combinations of other immunotherapies with monalizumab are currently enrolling patients, both in a curative and palliative setting (NCT06152523, NCT03801902: ARCHON-1 Trial, NCT05221840, NCT03794544, NCT05061550).
Other NKG2A-targeted mAbs are currently under development in first-line mNSCLC phase II trials: S095029, a human Fc-attenuated IgG1 mAb that binds NKG2A with an nM-range affinity [225,226] (NCT06162572), and BMS-986315, a human aNKG2A mAb (NCT06094296).
Table 5. Published data from clinical trials with anti-NKG2a monoclonal antibodies in non-small-cell lung cancer.
Table 5. Published data from clinical trials with anti-NKG2a monoclonal antibodies in non-small-cell lung cancer.
aNKG2A mAbTypeStudyPhaseSettingNIntervention1st EndpointResults with aLAG-3Ref
MonalizumabHumanized IgG4NeoCOASTIIResectable, stage IA3-IIIA, neoadjuvant83Mona + durva (aPD-1)
vs. ole (aCD73) + durva
vs. danvatirsen + durva
vs. durva
MPR
  • MPR 30.0% with mona + durva vs. 11.1% with durva
  • pCR 10.0% with mona + durva vs. 3.7% with durva
  • Grade ≥ 3 TRAE 0% with mona + durva and 0% with durva
[219]
COAST IIUnresectable, stage III, after chemoradiation189Mona + durva
vs. ole + durva
vs. durva
ORR
  • ORR 40.3% with mona + durva, vs. 23.9% with durva
  • mPFS 19.8 mo with mona + durva vs. 7.3 mo with durva (HR 0.63, 95% CI 0.40, 0.99)
  • mOS NR with mona + durva vs. 40.9 mo with durva (HR 0.77, 95% CI 0.44, 1.33)
  • Grade ≥ 3 TEAE 32.8% with mona + durva vs. 34.8% with durva
[220,221]
Patel et al.I/IIMetastatic, 2nd or 3rd line, ICI-naive20 with NSCLCMona + durvaSafety
  • ORR 10%
  • mPFS 1.9 mo
  • mOS 8.8 mo
  • Grade ≥ 3 TRAE 20% for NSCLC patients
[223]
PIONeeRIb/IIAMetastatic, 2nd or 3rd line, after aPD-(L)1114Mona + durva
vs. ole + durva
vs. ceralasertib + durva
vs. savolitinib + durva
vs. docetaxel
12 w DCR
  • 12 w DCR 24.1% with mona + durva vs. 54.5% with docetaxel
  • No safety data
[224]
mAb: monoclonal antibody; N: number of patients included; mona: monalizumab; durva: durvalumab; ole: oleclumab; MPR: major pathological response; pCR: pathological complete response; TRAE: treatment-related adverse events; ORR: objective response rate; mPFS: median progression-free survival; mo: months; HR: hazard ratio; CI: confidence index; mOS: median overall survival; TEAE: treatment-emergent adverse events; ICI: immune checkpoint inhibitor; 12 w DCR: 12-week disease control rate.

8. Targeting the CD39/CD73/Adenosine Pathway

CD39 (ENTPD1) and CD73 (ecto-5′-nucleotidase) are key enzymes in the extracellular adenosine pathway and play a critical role in modulating the TME (reviewed in references [227,228]). CD39 hydrolyzes adenosine triphosphate (ATP) and adenosine diphosphate (ADP) into adenosine monophosphate (AMP), while CD73 subsequently converts AMP to adenosine (ADO). Adenosine accumulation in the TME leads to potent immunosuppressive effects, aiding tumor progression and immune evasion [229]. CD73 is expressed on approximately 75% of B cells, 50% of CD8+ T cells, 10% of CD4+ T cells, and 2–5% of NK cells [230]. B cells, Tregs, Th17 cells, NK cells, myeloid-derived suppressor cells (MDSC), endothelial cells, fibroblasts, and stromal and tumor cells can co-express CD39 and CD73. The overexpression of CD39 and CD73 is associated with worse prognoses in various cancers, including melanoma, lung cancer, colorectal cancer, and breast cancer [231]. Recent insights into their immunoregulatory roles have positioned them as promising targets for next-generation cancer immunotherapies.
Multiple mechanisms could explain the deleterious effect of the CD39/CD73 pathway on the antitumor immune response (Figure 6). Extracellular ATP (eATP), released by stressed or dying cells in the TME, could promote inflammation and immune activation via purinergic receptors like P2X7. CD39 and CD73 convert eATP into adenosine, dampening immune responses by binding adenosine receptors (A2A and A2B). This inhibits T cell proliferation, cytokine production, and effector functions while enhancing Treg activity. Adenosine signaling directly suppresses cytotoxic CD8+ T cells and NK cells, while promoting immune checkpoints like PD-1 and CTLA-4. CD39 and CD73 also enhance angiogenesis and metastasis by increasing VEGF and IL-8 production in the TME. CD39/CD73 activity skews macrophages from a pro-inflammatory M1 phenotype to an immunosuppressive M2 phenotype and could also impair DC maturation, reduce antigen presentation, and suppress pro-inflammatory cytokine production [232]. Published results from clinical trials with drugs targeting the CD39/CD73/adenosine pathway are summarized in Table 6.

8.1. CD39 Targeting

CD39’s enzymatic activity reduces eATP and converts ATP into ADP. High CD39 expression is observed in terminally exhausted T cells, notably tumor-specific exhausted T cells. Preclinical studies have shown that blocking CD39 mAbs or using inhibitors could impede tumor growth and improve CD8 antitumor immune response in various murine models of melanoma, lung carcinoma, and colon tumors. Combining CD39-blocking mAbs with aPD-1 showed synergistic effects, further enhancing tumor control and immune response, thus suggesting a rationale for testing this combination in patients.

IPH5201

IPH5201 is an aCD39 mAb that has been assessed in a phase I basket trial enrolling patients with advanced solid tumors in combination with durvalumab [233]. The combination reached a median PFS and OS of 1.4 and 8.2 months, respectively, but data have not been published yet, particularly regarding the mNSCLC population. The phase II MATISSE [234] trial is currently assessing this combination in a multicenter, single-arm study, combining neoadjuvant IPH5201, durvalumab, and platinum-doublet chemotherapy followed by surgery then 1-year of adjuvant IH5201 and durvalumab, in patients with resectable, early-stage (II to IIIA) NSCLC.
Other CD39-targeted mAbs have been evaluated in early-phase basket trials in advanced solid tumors: SRF617 [226] (NCT05177770), TTX-030 [235] (NCT06119217 for pancreatic cancer, NCT03884556, NCT04306900), and ES002023 (NCT05075564).

8.2. CD73 Targeting

CD73’s enzymatic activity transforms AMP into adenosine, a powerful immunosuppressive enzyme. aCD73 mAbs were shown to enhance pro-mitogenic effects of anti-CD3 mAbs on human T cells in vitro [236]. In animals, CD73-deficient mice also have increased antitumor immunity, and preclinical studies have shown synergy between aCD73 therapy and immunotherapy [237,238]. Targeting CD73 has also been shown to suppress tumorigenesis by affecting anti-apoptotic proteins [239] or suppressing tumor angiogenesis [240].

Oleclumab

Oleclumab is a first-in-class therapeutic aCD73 mAb, a human IgG1λ, tested in phase I alone or with durvalumab with no safety concerns [241]. Regarding the neoadjuvant setting, in the phase II NeoCOAST platform trial [219], 24 patients received a single cycle of oleclumab with durvalumab before surgery for stage IA3-IIIA NSCLC. With oleclumab on top of aPD-L1, the MPR rate increased from 11.1% to 19.0% and the pCR rate from 3.7 to 9.5%. In a more advanced setting, the phase II COAST study [220,221] (unresectable stage III NSCLC) assessed the addition of oleclumab to adjuvant durvalumab after chemoradiation. The addition of oleclumab to durvalumab increased the ORR from 23.9% with durvalumab monotherapy to 35.0% with the combination and prolonged mPFS from 7.3 months with durvalumab monotherapy to 21.1 months. The combination might also prolong OS (data not mature). The phase Ib/IIA PIONeeR study [224] tested durvalumab and oleclumab therapy but with mNSCLC patients who had already experienced disease progression after sequential or concomitant PD-(L)1 and platinum-based chemotherapy. The 12-week DCR reached with the combination was 0% vs. 54.5% with docetaxel, suggesting lower efficacy in this context, as with the monalizumab and durvalumab combination.

8.3. Adenosine Receptor Inhibitors

An alternative strategy to target adenosine pathways is to target adenosine receptors. Extracellular adenosine can activate four distinct P1 purinergic receptors, namely A1, A2a, A2b, and A3 adenosine receptors [242]. A1, A2a, and A3 are considered as high-affinity receptors for adenosine, while the A2b receptor has lower affinity. The A2a and A2b receptor antagonists demonstrated safety and clinical activity in solid tumors when combined with chemotherapy/immunotherapy.

8.3.1. Etrumadenant

Etrumadenant is an orally bioavailable, selective, A2a and A2b receptor antagonist that has demonstrated safety and clinical activity in solid tumors when combined with chemo/immunotherapy. The ARC-7 [108] phase II trial (NCT04262856) contains an arm with the combination of zimberelimab, domvanalimab, and etrumadenant in treatment-naive patients with PDL1 > 50% (TPS) and no targetable oncogenic addiction. With the addition of etrumadenant and domvanalimab to the aPD-1, the ORR increased from 12% to 18%, and the median PFS increased from 5.4 to 10.9 months.

8.3.2. Taminadenant

Taminadenant is an orally bioavailable, A2a receptor antagonist. In a phase I/Ib study [243], mNSCLC patients treated with at least one prior therapy received taminadenant with or without spartalizumab (aPD-1). The ORRs with the combination of taminadenant plus aPD-1 and with taminadenant monotherapy were 8.3% and 9.5%, respectively. In the taminadenant monotherapy arm, the median PFS and OS were 3.9 and 9.7 months, respectively, compared to 2.8 and 5.4 months, in the combination arm. These data suggest that targeting the adenosine pathway at various sites of the CD39/CD73/adenosine receptor axis might be a promising approach to enhance the antitumor immune response in the setting of immunotherapy combinations. Translational studies are needed to determine key biomarkers for selecting mNSCLC patients who would be more likely to respond to such therapies.
Table 6. Published data from clinical trials with drugs targeting the CD39/CD73/adenosine pathway.
Table 6. Published data from clinical trials with drugs targeting the CD39/CD73/adenosine pathway.
aCD73 mAbTypeStudyPhaseSettingNIntervention1st EndpointResults with aCD73Ref
OleclumabHuman IgG1λNeoCOASTIIResectable, stage IA3-IIIA, neoadjuvant83Mona (aNKG2A) + durva (aPD-L1)
vs. ole + durva
vs. danvatirsen + durva
vs. durva
MPR
  • MPR 19.0% with ole + durva vs. 11.1% with durva
  • pCR 9.5% with ole + durva vs. 3.7% with durva
  • Grade ≥ 3 TRAE 4.8% with ole + durva and 0% with durva
[219]
COAST IIUnresectable, stage III, after chemoradiation189Mona + durva
vs. ole + durva
vs. durva
ORR
  • ORR 35.0% with ole + durva, vs. 23.9% with durva
  • mPFS 21.1 mo with ole + durva vs. 7.3 mo with durva (HR 0.59, 95% CI 0.37, 0.93)
  • mOS NR with ole + durva vs. 40.9 mo with durva (HR 0.69, 95% CI 0.40, 1.20)
  • Grade ≥ 3 TEAE 33.9% with ole + durva vs. 34.8% with durva
[220,221]
PIONeeRIb/IIAMetastatic, 2nd or 3rd line, after aPD-(L)1114Mona + durva
vs. ole + durva
vs. ceralasertib + durva
vs. savolitinib + durva
vs. docetaxel
12 w DCR
  • 12 w DCR 0% with ole + durva (prematurely closed due to lack of efficacy) vs. was 54.5% with docetaxel
  • No safety data
[224]
Adenosine R ITypeStudyPhaseSettingNIntervention1st EndpointResults with Adenosine R IRef
EtrumadenantSelective dual antagonist of A2a and A2bARC-7IIMetastatic, 1st line, PD-L1 ≥50% 151Domva (aTIGIT) + zimbe (aPD-1) + etrumadenant
vs. domva + zimbe
vs. zimbe
ORR, PFS
  • ORR 18% with domva + zimbe + etrumadenant vs. 12% with zimbe
  • mPFS 10.9 mo with domva + zimbe + etrumadenant vs. 5.4 mo with zimbe (HR 0.65, 95% CI 0.37–1.1)
  • Grade ≥ 3 TEAE 52% with domva + zimbe + etrumadenant vs. 58% with zimbe
[108]
TaminadenantA2a antagonistChiappori et al.I/IbMetastatic, ≥2nd line50Taminadenant
vs. taminadenant + sparta (aPD-1)
Safety
  • ORR 9.5% with taminadenant vs. 8.3% with taminadenant + sparta
  • mPFS 3.9 mo with taminadenant vs. 2.8 mo with taminadenant + sparta
  • mOS 9.7 mo with taminadenant vs. 5.4 mo with taminadenant + sparta
  • Grade ≥ 3 TRAE 16.0% with taminadenant vs. 36.0% with taminadenant + sparta
[243]
mAb: monoclonal antibody; N: number of patients included; mona: monalizumab; durva: durvalumab; ole: oleclumab; MPR: major pathological response; pCR: pathological complete response; TRAE: treatment-related adverse events; ORR: objective response rate; mPFS: median progression-free survival; mo: months; HR: hazard ratio; CI: confidence index; mOS: median overall survival; TEAE: treatment-emergent adverse events; 12 w DCR: 12-week disease control rate; adenosine R I: adenosine receptor inhibitor; domva: domvanalimab; zimbe: zimberelimab; PFS: progression-free survival; sparta: spartalizumab.

9. Conclusions

Immunotherapy has transformed the landscape of NSCLC treatment, yet the limitations of PD-1/PD-L1 targeting underscore the need for novel strategies. Emerging immune checkpoints, such as CTLA-4, TIGIT, LAG-3, and TIM-3, provide promising avenues to overcome resistance and enhance antitumor immunity. Advances in bispecific and Fc-engineered antibodies offer additional potential for therapeutic synergy and precision. Going forward, a deeper understanding of the tumor microenvironment, alongside refined biomarkers for patient selection, will be crucial in optimizing these approaches and expanding their impact on clinical outcomes.

Author Contributions

N.R. and F.G. contributed to the writing of the manuscript; N.R. provided figures and tables; N.R., C.K., and F.G contributed to the proofreading and corrections of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

We wish to thank Fiona Ecarnot (EA3920, University of Franche-Comté, Besançon, France) for English correction and helpful comments.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Hendriks, L.E.L.; Remon, J.; Faivre-Finn, C.; Garassino, M.C.; Heymach, J.V.; Kerr, K.M.; Tan, D.S.W.; Veronesi, G.; Reck, M. Non-small-cell lung cancer. Nat. Rev. Dis. Primers 2024, 10, 71. [Google Scholar] [CrossRef] [PubMed]
  2. Meyer, M.L.; Fitzgerald, B.G.; Paz-Ares, L.; Cappuzzo, F.; Jänne, P.A.; Peters, S.; Hirsch, F.R. New promises and challenges in the treatment of advanced non-small-cell lung cancer. Lancet 2024, 404, 803–822. [Google Scholar] [CrossRef] [PubMed]
  3. Waldman, A.D.; Fritz, J.M.; Lenardo, M.J. A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat. Rev. Immunol. 2020, 20, 651–668. [Google Scholar] [CrossRef] [PubMed]
  4. Hendriks, L.E.; Kerr, K.M.; Menis, J.; Mok, T.S.; Nestle, U.; Passaro, A.; Peters, S.; Planchard, D.; Smit, E.F.; Solomon, B.J.; et al. Non-oncogene-addicted metastatic non-small-cell lung cancer: ESMO clinical practice guideline for diagnosis, treatment and follow-up. Ann. Oncol. 2023, 34, 358–376. [Google Scholar] [CrossRef]
  5. Garon, E.B.; Rizvi, N.A.; Hui, R.; Leighl, N.; Balmanoukian, A.S.; Eder, J.P.; Patnaik, A.; Aggarwal, C.; Gubens, M.; Horn, L.; et al. Pembrolizumab for the treatment of non–small-cell lung cancer. N. Engl. J. Med. 2015, 372, 2018–2028. [Google Scholar] [CrossRef]
  6. Borghaei, H.; Paz-Ares, L.; Horn, L.; Spigel, D.R.; Steins, M.; Ready, N.E.; Chow, L.Q.; Vokes, E.E.; Felip, E.; Holgado, E.; et al. Nivolumab versus Docetaxel in advanced Nonsquamous non–small-cell lung cancer. N. Engl. J. Med. 2015, 373, 1627–1639. [Google Scholar] [CrossRef]
  7. Reck, M.; Rodríguez-Abreu, D.; Robinson, A.G.; Hui, R.; Csőszi, T.; Fülöp, A.; Gottfried, M.; Peled, N.; Tafreshi, A.; Cuffe, S.; et al. Pembrolizumab versus Chemotherapy for PD-L1–positive non–small-cell lung cancer. N. Engl. J. Med. 2016, 375, 1823–1833. [Google Scholar] [CrossRef]
  8. Gandhi, L.; Rodríguez-Abreu, D.; Gadgeel, S.; Esteban, E.; Felip, E.; De Angelis, F.; Domine, M.; Clingan, P.; Hochmair, M.J.; Powell, S.F.; et al. Pembrolizumab plus Chemotherapy in metastatic non–small-cell lung cancer. N. Engl. J. Med 2018, 378, 2078–2092. [Google Scholar] [CrossRef]
  9. Garassino, M.C.; Gadgeel, S.; Speranza, G.; Felip, E.; Esteban, E.; Dómine, M.; Hochmair, M.J.; Powell, S.F.; Bischoff, H.G.; Peled, N.; et al. Pembrolizumab plus Pemetrexed and Platinum in Nonsquamous non–small-cell lung cancer: 5-year outcomes from the phase 3 KEYNOTE-189 study. J. Clin. Oncol. 2023, 41, 1992–1998. [Google Scholar] [CrossRef]
  10. Chen, L.; Han, X. Anti–PD-1/PD-L1 therapy of human cancer: Past, present, and future. J. Clin. Investig. 2015, 125, 3384–3391. [Google Scholar] [CrossRef]
  11. Sharpe, A.H.; Pauken, K.E. The diverse functions of the PD1 inhibitory pathway. Nat. Rev. Immunol. 2018, 18, 153–167. [Google Scholar] [CrossRef]
  12. Jiang, Y.; Chen, M.; Nie, H.; Yuan, Y. PD-1 and PD-L1 in cancer immunotherapy: Clinical implications and future considerations. Human Vaccines Immunother. 2019, 15, 1111–1122. [Google Scholar] [CrossRef] [PubMed]
  13. Chamoto, K.; Yaguchi, T.; Tajima, M.; Honjo, T. Insights from a 30-year journey: Function, regulation and therapeutic modulation of PD1. Nat. Rev. Immunol 2023, 23, 682–695. [Google Scholar] [CrossRef] [PubMed]
  14. Blank, C.U.; Haining, W.N.; Held, W.; Hogan, P.G.; Kallies, A.; Lugli, E.; Lynn, R.C.; Philip, M.; Rao, A.; Restifo, N.P.; et al. Defining ‘T Cell Exhaustion’. Nat. Rev. Immunol. 2019, 19, 665–674. [Google Scholar] [CrossRef] [PubMed]
  15. Baessler, A.; Vignali, D.A.A. T Cell exhaustion. Annu. Rev. Immunol. 2024, 42, 179–206. [Google Scholar] [CrossRef]
  16. Alfei, F.; Kanev, K.; Hofmann, M.; Wu, M.; Ghoneim, H.E.; Roelli, P.; Utzschneider, D.T.; Von Hoesslin, M.; Cullen, J.G.; Fan, Y.; et al. TOX reinforces the phenotype and longevity of exhausted T cells in chronic viral infection. Nature 2019, 571, 265–269. [Google Scholar] [CrossRef]
  17. Khan, O.; Giles, J.R.; McDonald, S.; Manne, S.; Ngiow, S.F.; Patel, K.P.; Werner, M.T.; Huang, A.C.; Alexander, K.A.; Wu, J.E.; et al. TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion. Nature 2019, 571, 211–218. [Google Scholar] [CrossRef]
  18. Scott, A.C.; Dündar, F.; Zumbo, P.; Chandran, S.S.; Klebanoff, C.A.; Shakiba, M.; Trivedi, P.; Menocal, L.; Appleby, H.; Camara, S.; et al. TOX is a critical regulator of tumour-specific T cell differentiation. Nature 2019, 571, 270–274. [Google Scholar] [CrossRef]
  19. Yao, C.; Sun, H.W.; Lacey, N.E.; Ji, Y.; Moseman, E.A.; Shih, H.Y.; Heuston, E.F.; Kirby, M.; Anderson, S.; Cheng, J.; et al. Single-cell RNA-Seq reveals TOX as a key regulator of CD8+ T cell persistence in chronic infection. Nat. Immunol. 2019, 20, 890–901. [Google Scholar] [CrossRef]
  20. Yu, H.; Boyle, T.A.; Zhou, C.; Rimm, D.L.; Hirsch, F.R. PD-L1 expression in lung cancer. J. Thorac. Oncol. 2016, 11, 964–975. [Google Scholar] [CrossRef]
  21. Greillier, L.; Tomasini, P.; Barlesi, F. The clinical utility of tumor mutational burden in non-small cell lung cancer. Transl. Lung Cancer Res. 2018, 7, 639–646. [Google Scholar] [CrossRef] [PubMed]
  22. Marabelle, A.; Fakih, M.; Lopez, J.; Shah, M.; Shapira-Frommer, R.; Nakagawa, K.; Chung, H.C.; Kindler, H.L.; Lopez-Martin, J.A.; Miller, W.H.; et al. Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: Prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol. 2020, 21, 1353–1365. [Google Scholar] [CrossRef] [PubMed]
  23. Ricciuti, B.; Wang, X.; Alessi, J.V.; Rizvi, H.; Mahadevan, N.R.; Li, Y.Y.; Polio, A.; Lindsay, J.; Umeton, R.; Sinha, R.; et al. Association of high tumor mutation burden in non–small cell lung cancers with increased immune infiltration and improved clinical outcomes of PD-L1 blockade across PD-L1 expression levels. JAMA Oncol. 2022, 8, 1160. [Google Scholar] [CrossRef]
  24. Ayers, M.; Lunceford, J.; Nebozhyn, M.; Murphy, E.; Loboda, A.; Kaufman, D.R.; Albright, A.; Cheng, J.D.; Kang, S.P.; Shankaran, V.; et al. IFN-γ–related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Investig. 2017, 127, 2930–2940. [Google Scholar] [CrossRef] [PubMed]
  25. Cristescu, R.; Mogg, R.; Ayers, M.; Albright, A.; Murphy, E.; Yearley, J.; Sher, X.; Liu, X.Q.; Lu, H.; Nebozhyn, M.; et al. Pan-tumor genomic biomarkers for PD-1 checkpoint blockade–based immunotherapy. Science 2018, 362, eaar3593. [Google Scholar] [CrossRef]
  26. Ott, P.A.; Bang, Y.J.; Piha-Paul, S.A.; Razak, A.R.A.; Bennouna, J.; Soria, J.C.; Rugo, H.S.; Cohen, R.B.; O’Neil, B.H.; Mehnert, J.M.; et al. T-cell–inflamed gene-expression profile, programmed death ligand 1 expression, and tumor mutational burden predict efficacy in patients treated with pembrolizumab across 20 cancers: KEYNOTE-028. J. Clin. Oncol. 2019, 37, 318–327. [Google Scholar] [CrossRef]
  27. Ghiringhelli, F.; Bibeau, F.; Greillier, L.; Fumet, J.D.; Ilie, A.; Monville, F.; Laugé, C.; Catteau, A.; Boquet, I.; Majdi, A.; et al. Immunoscore immune checkpoint using spatial quantitative analysis of CD8 and PD-L1 markers is predictive of the efficacy of anti- PD1/PD-L1 immunotherapy in non-small cell lung cancer. Ebiomedicine 2023, 92, 104633. [Google Scholar] [CrossRef] [PubMed]
  28. Hashimoto, M.; Kamphorst, A.O.; Im, S.J.; Kissick, H.T.; Pillai, R.N.; Ramalingam, S.S.; Araki, K.; Ahmed, R. CD8 T cell exhaustion in chronic infection and cancer: Opportunities for interventions. Annu. Rev. Med. 2018, 69, 301–318. [Google Scholar] [CrossRef]
  29. McLane, L.M.; Abdel-Hakeem, M.S.; Wherry, E.J. CD8 T cell exhaustion during chronic viral infection and cancer. Annu. Rev. Immunol. 2019, 37, 457–495. [Google Scholar] [CrossRef]
  30. Zhao, X.; Shan, Q.; Xue, H.H. TCF1 in T cell immunity: A broadened frontier. Nat. Rev. Immunol. 2022, 22, 147–157. [Google Scholar] [CrossRef]
  31. Bordon, Y. TOX for tired T cells. Nat. Rev. Immunol. 2019, 19, 476. [Google Scholar] [CrossRef]
  32. Blackburn, S.D.; Shin, H.; Freeman, G.J.; Wherry, E.J. Selective expansion of a subset of exhausted CD8 T cells by αPD-L1 Blockade. Proc. Natl. Acad. Sci. USA 2008, 105, 15016–15021. [Google Scholar] [CrossRef] [PubMed]
  33. Utzschneider, D.T.; Charmoy, M.; Chennupati, V.; Pousse, L.; Ferreira, D.P.; Calderon-Copete, S.; Danilo, M.; Alfei, F.; Hofmann, M.; Wieland, D.; et al. T cell factor 1-expressing memory-like CD8+ T cells sustain the immune response to chronic viral infections. Immunity 2016, 45, 415–427. [Google Scholar] [CrossRef] [PubMed]
  34. Im, S.J.; Hashimoto, M.; Gerner, M.Y.; Lee, J.; Kissick, H.T.; Burger, M.C.; Shan, Q.; Hale, J.S.; Lee, J.; Nasti, T.H.; et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 2016, 537, 417–421. [Google Scholar] [CrossRef]
  35. Pauken, K.E.; Sammons, M.A.; Odorizzi, P.M.; Manne, S.; Godec, J.; Khan, O.; Drake, A.M.; Chen, Z.; Sen, D.R.; Kurachi, M.; et al. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science 2016, 354, 1160–1165. [Google Scholar] [CrossRef] [PubMed]
  36. Sen, D.R.; Kaminski, J.; Barnitz, R.A.; Kurachi, M.; Gerdemann, U.; Yates, K.B.; Tsao, H.W.; Godec, J.; LaFleur, M.W.; Brown, F.D.; et al. The epigenetic landscape of T cell exhaustion. Science 2016, 354, 1165–1169. [Google Scholar] [CrossRef]
  37. Belk, J.A.; Daniel, B.; Satpathy, A.T. Epigenetic regulation of T cell exhaustion. Nat. Immunol. 2022, 23, 848–860. [Google Scholar] [CrossRef]
  38. Giles, J.R.; Ngiow, S.F.; Manne, S.; Baxter, A.E.; Khan, O.; Wang, P.; Staupe, R.; Abdel-Hakeem, M.S.; Huang, H.; Mathew, D.; et al. Shared and distinct biological circuits in effector, memory and exhausted CD8+ T cells revealed by temporal single-cell transcriptomics and epigenetics. Nat. Immunol. 2022, 23, 1600–1613. [Google Scholar] [CrossRef]
  39. Andrews, L.P.; Butler, S.C.; Cui, J.; Cillo, A.R.; Cardello, C.; Liu, C.; Brunazzi, E.A.; Baessler, A.; Xie, B.; Kunning, S.R.; et al. LAG-3 and PD-1 synergize on CD8+ T cells to drive t cell exhaustion and hinder autocrine IFN-γ-dependent anti-tumor immunity. Cell 2024, 187, 4355–4372.e22. [Google Scholar] [CrossRef]
  40. Cillo, A.R.; Cardello, C.; Shan, F.; Karapetyan, L.; Kunning, S.; Sander, C.; Rush, E.; Karunamurthy, A.; Massa, R.C.; Rohatgi, A.; et al. Blockade of LAG-3 and PD-1 leads to co-expression of cytotoxic and exhaustion gene modules in CD8+ T cells to promote antitumor immunity. Cell 2024, 187, 4373–4388.e15. [Google Scholar] [CrossRef]
  41. Hofmann, M.; Thimme, R.; Schamel, W.W. PD-1 and LAG-3: Synergistic fostering of T cell exhaustion. Sig. Transduct. Target Ther. 2024, 9, 291. [Google Scholar] [CrossRef] [PubMed]
  42. Ngiow, S.F.; Manne, S.; Huang, Y.J.; Azar, T.; Chen, Z.; Mathew, D.; Chen, Q.; Khan, O.; Wu, J.E.; Alcalde, V.; et al. LAG-3 sustains TOX expression and regulates the CD94/NKG2-Qa-1b axis to govern exhausted CD8 T Cell NK receptor expression and cytotoxicity. Cell 2024, 187, 4336–4354.e19. [Google Scholar] [CrossRef] [PubMed]
  43. Tawbi, H.A.; Schadendorf, D.; Lipson, E.J.; Ascierto, P.A.; Matamala, L.; Castillo Gutiérrez, E.; Rutkowski, P.; Gogas, H.J.; Lao, C.D.; De Menezes, J.J.; et al. Relatlimab and nivolumab versus nivolumab in untreated advanced melanoma. N. Engl. J. Med. 2022, 386, 24–34. [Google Scholar] [CrossRef]
  44. Morgensztern, D.; Chaudhry, A.; Iannotti, N.; Acevedo, A.; Balaburski, G.; Balogh, A.; Peters, S. 1359TiP RELATIVITY-104: First-Line Relatlimab (RELA) + Nivolumab (NIVO) with chemotherapy vs nivo with chemotherapy in stage IV or recurrent Non-Small Cell Lung Cancer (NSCLC): A phase II, randomized, double-blind study. Ann. Oncol. 2021, 32, S1030. [Google Scholar] [CrossRef]
  45. Schuler, M.; Cuppens, K.; Plönes, T.; Wiesweg, M.; Du Pont, B.; Hegedus, B.; Köster, J.; Mairinger, F.; Darwiche, K.; Paschen, A.; et al. Neoadjuvant nivolumab with or without relatlimab in resectable non-small-cell lung cancer: A randomized phase 2 trial. Nat. Med. 2024, 30, 1602–1611. [Google Scholar] [CrossRef]
  46. Augustin, R.C.; Bao, R.; Luke, J.J. Targeting Cbl-b in cancer immunotherapy. J. Immunother. Cancer 2023, 11, e006007. [Google Scholar] [CrossRef]
  47. Chocarro, L.; Blanco, E.; Fernandez-Rubio, L.; Garnica, M.; Zuazo, M.; Garcia, M.J.; Bocanegra, A.; Echaide, M.; Johnston, C.; Edwards, C.J.; et al. PD-1/LAG-3 Co-signaling profiling uncovers CBL ubiquitin ligases as key immunotherapy targets. EMBO Mol. Med. 2024, 16, 1791–1816. [Google Scholar] [CrossRef] [PubMed]
  48. Chu, Y.; Dai, E.; Li, Y.; Han, G.; Pei, G.; Ingram, D.R.; Thakkar, K.; Qin, J.J.; Dang, M.; Le, X.; et al. Pan-cancer T cell atlas links a cellular stress response state to immunotherapy resistance. Nat. Med. 2023, 29, 1550–1562. [Google Scholar] [CrossRef] [PubMed]
  49. Globig, A.M.; Zhao, S.; Roginsky, J.; Maltez, V.I.; Guiza, J.; Avina-Ochoa, N.; Heeg, M.; Araujo Hoffmann, F.; Chaudhary, O.; Wang, J.; et al. The Β1-adrenergic receptor links sympathetic nerves to T cell exhaustion. Nature 2023, 622, 383–392. [Google Scholar] [CrossRef]
  50. Baek, A.E. Stress exhausts T cells. Sci. Signal. 2023, 16, eadl0724. [Google Scholar] [CrossRef]
  51. Zhang, J.; Li, J.; Hou, Y.; Lin, Y.; Zhao, H.; Shi, Y.; Chen, K.; Nian, C.; Tang, J.; Pan, L.; et al. Osr2 functions as a biomechanical checkpoint to aggravate CD8+ T cell exhaustion in tumor. Cell 2024, 187, 3409–3426.e24. [Google Scholar] [CrossRef] [PubMed]
  52. Krummel, M.F.; Allison, J.P. CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation. J. Exp. Med. 1995, 182, 459–465. [Google Scholar] [CrossRef] [PubMed]
  53. McCoy, K.D.; Le Gros, G. The role of CTLA-4 in the regulation of T cell immune responses. Immunol. Cell Biol. 1999, 77, 1–10. [Google Scholar] [CrossRef]
  54. Sobhani, N.; Tardiel-Cyril, D.R.; Davtyan, A.; Generali, D.; Roudi, R.; Li, Y. CTLA-4 in regulatory T cells for cancer immunotherapy. Cancers 2021, 13, 1440. [Google Scholar] [CrossRef]
  55. Walker, L.S.K.; Sansom, D.M. The emerging role of CTLA4 as a cell-extrinsic regulator of T cell responses. Na.t Rev. Immunol. 2011, 11, 852–863. [Google Scholar] [CrossRef]
  56. Hodi, F.S.; O’Day, S.J.; McDermott, D.F.; Weber, R.W.; Sosman, J.A.; Haanen, J.B.; Gonzalez, R.; Robert, C.; Schadendorf, D.; Hassel, J.C.; et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 2010, 363, 711–723. [Google Scholar] [CrossRef]
  57. Sharma, P.; Goswami, S.; Raychaudhuri, D.; Siddiqui, B.A.; Singh, P.; Nagarajan, A.; Liu, J.; Subudhi, S.K.; Poon, C.; Gant, K.L.; et al. Immune checkpoint therapy—Current perspectives and future directions. Cell 2023, 186, 1652–1669. [Google Scholar] [CrossRef] [PubMed]
  58. Quezada, S.A. CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells. J. Clin. Investig. 2006, 116, 1935–1945. [Google Scholar] [CrossRef]
  59. Lax, B.M.; Palmeri, J.R.; Lutz, E.A.; Sheen, A.; Stinson, J.A.; Duhamel, L.; Santollani, L.; Kennedy, A.; Rothschilds, A.M.; Spranger, S.; et al. Both intratumoral regulatory T cell depletion and CTLA-4 antagonism are required for maximum efficacy of anti-CTLA-4 antibodies. Proc. Natl. Acad. Sci. USA 2023, 120, e2300895120. [Google Scholar] [CrossRef]
  60. Tang, F.; Du, X.; Liu, M.; Zheng, P.; Liu, Y. Anti-CTLA-4 antibodies in cancer immunotherapy: Selective depletion of intratumoral regulatory T cells or checkpoint blockade? Cell Biosci. 2018, 8, 30. [Google Scholar] [CrossRef]
  61. Sato, Y.; Casson, C.N.; Matsuda, A.; Kim, J.I.; Shi, J.Q.; Iwasaki, S.; Chen, S.; Modrell, B.; Chan, C.; Tavares, D.; et al. Fc-independent functions of anti-CTLA-4 antibodies contribute to anti-tumor efficacy. Cancer Immunol. Immunother. 2022, 71, 2421–2431. [Google Scholar] [CrossRef] [PubMed]
  62. Robert, L.; Tsoi, J.; Wang, X.; Emerson, R.; Homet, B.; Chodon, T.; Mok, S.; Huang, R.R.; Cochran, A.J.; Comin-Anduix, B.; et al. CTLA4 blockade broadens the peripheral T-cell receptor repertoire. Clin. Cancer Res. 2014, 20, 2424–2432. [Google Scholar] [CrossRef] [PubMed]
  63. Hellmann, M.D.; Paz-Ares, L.; Bernabe Caro, R.; Zurawski, B.; Kim, S.W.; Carcereny Costa, E.; Park, K.; Alexandru, A.; Lupinacci, L.; De La Mora Jimenez, E.; et al. Nivolumab plus ipilimumab in advanced non–small-cell lung cancer. N. Engl. J. Med. 2019, 381, 2020–2031. [Google Scholar] [CrossRef] [PubMed]
  64. Motzer, R.J.; Tannir, N.M.; McDermott, D.F.; Arén Frontera, O.; Melichar, B.; Choueiri, T.K.; Plimack, E.R.; Barthélémy, P.; Porta, C.; George, S.; et al. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N. Engl. J. Med. 2018, 378, 1277–1290. [Google Scholar] [CrossRef]
  65. Baas, P.; Scherpereel, A.; Nowak, A.K.; Fujimoto, N.; Peters, S.; Tsao, A.S.; Mansfield, A.S.; Popat, S.; Jahan, T.; Antonia, S.; et al. First-line nivolumab plus ipilimumab in unresectable malignant pleural mesothelioma (CheckMate 743): A multicentre, randomised, open-label, phase 3 trial. Lancet 2021, 397, 375–386. [Google Scholar] [CrossRef]
  66. Melero, I.; Yau, T.; Kang, Y.K.; Kim, T.Y.; Santoro, A.; Sangro, B.; Kudo, M.; Hou, M.M.; Matilla, A.; Tovoli, F.; et al. Nivolumab plus ipilimumab combination therapy in patients with advanced hepatocellular carcinoma previously treated with sorafenib: 5-year results from CheckMate 040. Ann. Oncol. 2024, 35, 537–548. [Google Scholar] [CrossRef]
  67. Andre, T.; Elez, E.; Van Cutsem, E.; Jensen, L.H.; Bennouna, J.; Mendez, G.; Schenker, M.; De La Fouchardiere, C.; Limon, M.L.; Yoshino, T.; et al. Nivolumab plus ipilimumab in microsatellite-instability–high metastatic colorectal cancer. N. Engl. J. Med. 2024, 391, 2014–2026. [Google Scholar] [CrossRef]
  68. Hossain, M.A. A Comprehensive review of immune checkpoint inhibitors for cancer treatment. Int. Immunopharmacol. 2024, 143, 113365. [Google Scholar] [CrossRef]
  69. Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N. Engl. J. Med. 2019, 381, 1535–1546. [Google Scholar] [CrossRef]
  70. Wolchok, J.D.; Chiarion-Sileni, V.; Rutkowski, P.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Queirolo, P.; Dummer, R.; Butler, M.O.; Hill, A.G.; et al. Final, 10-year outcomes with nivolumab plus ipilimumab in advanced melanoma. N. Engl. J. Med. 2025, 392, 11–22. [Google Scholar] [CrossRef]
  71. Paz-Ares, L.; Ciuleanu, T.E.; Cobo, M.; Schenker, M.; Zurawski, B.; Menezes, J.; Richardet, E.; Bennouna, J.; Felip, E.; Juan-Vidal, O.; et al. First-line nivolumab plus ipilimumab combined with two cycles of chemotherapy in patients with non-small-cell lung cancer (CheckMate 9LA): An international, randomised, open-label, phase 3 trial. Lancet Oncol. 2021, 22, 198–211. [Google Scholar] [CrossRef] [PubMed]
  72. Reck, M.; Ciuleanu, T.E.; Schenker, M.; Bordenave, S.; Cobo, M.; Juan-Vidal, O.; Reinmuth, N.; Richardet, E.; Felip, E.; Menezes, J.; et al. Five-year outcomes with first-line nivolumab plus ipilimumab with 2 cycles of chemotherapy versus 4 cycles of chemotherapy alone in patients with metastatic non-small cell lung cancer in the randomized CheckMate 9LA trial. Eur. J. Cancer 2024, 211, 114296. [Google Scholar] [CrossRef]
  73. Brahmer, J.R.; Lee, J.S.; Ciuleanu, T.E.; Bernabe Caro, R.; Nishio, M.; Urban, L.; Audigier-Valette, C.; Lupinacci, L.; Sangha, R.; Pluzanski, A.; et al. Five-year survival outcomes with nivolumab plus ipilimumab versus chemotherapy as first-line treatment for metastatic non–small-cell lung cancer in CheckMate 227. J. Clin. Oncol. 2023, 41, 1200–1212. [Google Scholar] [CrossRef] [PubMed]
  74. Ribas, A.; Kefford, R.; Marshall, M.A.; Punt, C.J.A.; Haanen, J.B.; Marmol, M.; Garbe, C.; Gogas, H.; Schachter, J.; Linette, G.; et al. Phase III randomized clinical trial comparing tremelimumab with standard-of-care chemotherapy in patients with advanced melanoma. J. Clin. Oncol. 2013, 31, 616–622. [Google Scholar] [CrossRef]
  75. Abou-Alfa, G.K.; Lau, G.; Kudo, M.; Chan, S.L.; Kelley, R.K.; Furuse, J.; Sukeepaisarnjaroen, W.; Kang, Y.K.; Van Dao, T.; De Toni, E.N.; et al. Tremelimumab plus durvalumab in unresectable hepatocellular carcinoma. NEJM Evid. 2022, 1, EVIDoa2100070. [Google Scholar] [CrossRef]
  76. Johnson, M.L.; Cho, B.C.; Luft, A.; Alatorre-Alexander, J.; Geater, S.L.; Laktionov, K.; Kim, S.W.; Ursol, G.; Hussein, M.; Lim, F.L.; et al. Durvalumab with or without tremelimumab in combination with chemotherapy as first-line therapy for metastatic non–small-cell lung cancer: The phase III POSEIDON study. J. Clin. Oncol. 2023, 41, 1213–1227. [Google Scholar] [CrossRef] [PubMed]
  77. Wang, S.J.; Dougan, S.K.; Dougan, M. Immune mechanisms of toxicity from checkpoint inhibitors. Trends Cancer 2023, 9, 543–553. [Google Scholar] [CrossRef]
  78. Lebbé, C.; Meyer, N.; Mortier, L.; Marquez-Rodas, I.; Robert, C.; Rutkowski, P.; Menzies, A.M.; Eigentler, T.; Ascierto, P.A.; Smylie, M.; et al. Evaluation of two dosing regimens for nivolumab in combination with ipilimumab in patients with advanced melanoma: Results from the phase IIIb/IV CheckMate 511 trial. J. Clin. Oncol. 2019, 37, 867–875. [Google Scholar] [CrossRef]
  79. O’Day, S.; Khoueiry, A.E.; Ramamurthy, C.; Bullock, A.; Shapiro, I.; Serina; Han, H.; Namagerdi, L.O.; Kaleta, R.; Wijatyk, A.; et al. 398 AGEN1181, an Fc engineered anti-CTLA-4 Antibody, Demonstrates Clinical Activity, Alone or in Combination with Balstilimab (Anti-PD-1), and Broadens the Therapeutic Potential of CTLA-4 Therapy. J. Immuno Ther. Cancer 2020, 8. [Google Scholar] [CrossRef]
  80. Chand, D.; Savitsky, D.A.; Krishnan, S.; Mednick, G.; Delepine, C.; Garcia-Broncano, P.; Soh, K.T.; Wu, W.; Wilkens, M.K.; Udartseva, O.; et al. Botensilimab, an Fc-enhanced anti–CTLA-4 antibody, is effective against tumors poorly responsive to conventional immunotherapy. Cancer Discov. 2024, 14, 2407–2429. [Google Scholar] [CrossRef]
  81. Bullock, A.J.; Schlechter, B.L.; Fakih, M.G.; Tsimberidou, A.M.; Grossman, J.E.; Gordon, M.S.; Wilky, B.A.; Pimentel, A.; Mahadevan, D.; Balmanoukian, A.S.; et al. Botensilimab plus balstilimab in relapsed/refractory microsatellite stable metastatic colorectal cancer: A phase 1 trial. Nat. Med. 2024, 30, 2558–2567. [Google Scholar] [CrossRef] [PubMed]
  82. Pang, X.; Huang, Z.; Zhong, T.; Zhang, P.; Wang, Z.M.; Xia, M.; Li, B. Cadonilimab, a tetravalent PD-1/CTLA-4 bispecific antibody with trans-binding and enhanced target binding avidity. Mabs 2023, 15, 2180794. [Google Scholar] [CrossRef] [PubMed]
  83. Zhao, Y.; Ma, Y.; Fan, Y.; Zhou, J.; Yang, N.; Yu, Q.; Zhuang, W.; Song, W.; Wang, Z.M.; Li, B.; et al. A multicenter, open-label phase Ib/II study of cadonilimab (anti PD-1 and CTLA-4 bispecific antibody) monotherapy in previously treated advanced non–small-cell lung cancer (AK104-202 Study). Lung Cancer 2023, 184, 107355. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, L.; Su, C.E.P. 11D.01 A phase II study of cadonilimab plus chemotherapy as first-line treatment for PD-L1-negative advanced non-small cell lung cancer: LungCadX. J. Thorac. Oncol. 2024, 19, S614. [Google Scholar] [CrossRef]
  85. Ma, Y.; Xue, J.; Zhao, Y.; Zhang, Y.; Huang, Y.; Yang, Y.; Fang, W.; Guo, Y.; Li, Q.; Ge, X.; et al. Phase I trial of KN046, a novel bispecific antibody targeting PD-L1 and CTLA-4 in patients with advanced solid tumors. J. Immunother. Cancer 2023, 11, e006654. [Google Scholar] [CrossRef]
  86. Zhao, Y.; Chen, G.; Li, X.; Wu, J.; Chang, B.; Hu, S.; Yang, S.; Xu, T.; Liu, Y.; Wang, N.; et al. KN046, a bispecific antibody against PD-L1 and CTLA-4, plus chemotherapy as first-line treatment for metastatic NSCLC: A multicenter phase 2 trial. Cell Rep. Med. 2024, 5, 101470. [Google Scholar] [CrossRef]
  87. Ahn, M.J.; Kim, S.W.; Costa, E.C.; Rodríguez, L.M.; Oliveira, J.; Insa Molla, M.A.; Majem, M.; Costa, L.; Su, W.C.; Lee, K.H.; et al. LBA56 MEDI5752 or Pembrolizumab (P) plus Carboplatin/Pemetrexed (CP) in treatment-Naïve (1L) non-small cell lung cancer (NSCLC): A phase Ib/II trial. Ann. Oncol. 2022, 33, S1423. [Google Scholar] [CrossRef]
  88. Spigel, D.R.; Ahn, M.J.; Majem, M.; Medina Rodríguez, L.; Lee, K.H.; Carcereny, E.; Hernández, A.A.; Insa, A.; Cho, E.K.; Besse, B.; et al. OA11.04 volrustomig + platinum doublet chemotherapy (CTx) in first-line non-small cell lung cancer (NSCLC): Phase 1b trial update. J. Thorac. Oncol. 2024, 19, S33–S34. [Google Scholar] [CrossRef]
  89. Johnson, M.L.; Arriola, E.; Kato, T.; Girard, N.; Gadgeel, S.M.; Wang, J.; Li, X.; Lowery, C.; Krug, L.M.; Ahn, M.J. eVOLVE-Lung02: A phase 3 study of first-line (1L) volrustomig plus chemotherapy (CT) versus pembrolizumab plus CT in metastatic non-small-cell lung cancer (mNSCLC) with PD-L1 tumor cell (TC) expression < 50%. J. Clin. Oncol. 2024, 42, TPS8652. [Google Scholar] [CrossRef]
  90. Sanchez-Correa, B.; Valhondo, I.; Hassouneh, F.; Lopez-Sejas, N.; Pera, A.; Bergua, J.M.; Arcos, M.J.; Bañas, H.; Casas-Avilés, I.; Durán, E.; et al. DNAM-1 and the TIGIT/PVRIG/TACTILE axis: Novel immune checkpoints for natural killer cell-based cancer immunotherapy. Cancers 2019, 11, 877. [Google Scholar] [CrossRef]
  91. Molfetta, R.; Zitti, B.; Lecce, M.; Milito, N.D.; Stabile, H.; Fionda, C.; Cippitelli, M.; Gismondi, A.; Santoni, A.; Paolini, R. CD155: A multi-functional molecule in tumor progression. Int. J. Mol. Sci. 2020, 21, 922. [Google Scholar] [CrossRef]
  92. Preillon, J.; Cuende, J.; Rabolli, V.; Garnero, L.; Mercier, M.; Wald, N.; Pappalardo, A.; Denies, S.; Jamart, D.; Michaux, A.C.; et al. Restoration of T-cell effector function, depletion of tregs, and direct killing of tumor cells: The multiple mechanisms of action of a-TIGIT antagonist antibodies. Mol. Cancer Ther. 2021, 20, 121–131. [Google Scholar] [CrossRef] [PubMed]
  93. Joller, N.; Lozano, E.; Burkett, P.R.; Patel, B.; Xiao, S.; Zhu, C.; Xia, J.; Tan, T.G.; Sefik, E.; Yajnik, V.; et al. Treg cells expressing the coinhibitory molecule TIGIT selectively inhibit proinflammatory Th1 and Th17 cell responses. Immunity 2014, 40, 569–581. [Google Scholar] [CrossRef]
  94. Harjunpää, H.; Guillerey, C. TIGIT as an emerging immune checkpoint. Clin. Exp. Immunol. 2020, 200, 108–119. [Google Scholar] [CrossRef] [PubMed]
  95. Joller, N.; Hafler, J.P.; Brynedal, B.; Kassam, N.; Spoerl, S.; Levin, S.D.; Sharpe, A.H.; Kuchroo, V.K. Cutting edge: TIGIT has T cell-intrinsic inhibitory functions. J. Immunol. 2011, 186, 1338–1342. [Google Scholar] [CrossRef] [PubMed]
  96. Li, M.; Xia, P.; Du, Y.; Liu, S.; Huang, G.; Chen, J.; Zhang, H.; Hou, N.; Cheng, X.; Zhou, L.; et al. T-cell immunoglobulin and ITIM domain (TIGIT) receptor/poliovirus receptor (PVR) ligand engagement suppresses interferon—γ production of natural killer cells via β—Arrestin 2-mediated negative signaling. J. Biol. Chem. 2014, 289, 17647–17657. [Google Scholar] [CrossRef]
  97. Johnston, R.J.; Comps-Agrar, L.; Hackney, J.; Yu, X.; Huseni, M.; Yang, Y.; Park, S.; Javinal, V.; Chiu, H.; Irving, B.; et al. The immunoreceptor TIGIT regulates antitumor and antiviral CD8 + T cell effector function. Cancer Cell 2014, 26, 923–937. [Google Scholar] [CrossRef]
  98. Yu, X.; Harden, K.; Gonzalez, L.C.; Francesco, M.; Chiang, E.; Irving, B.; Tom, I.; Ivelja, S.; Refino, C.J.; Clark, H.; et al. The surface protein TIGIT suppresses T cell activation by promoting the generation of mature immunoregulatory dendritic cells. Nat. Immunol. 2009, 10, 48–57. [Google Scholar] [CrossRef]
  99. Lepletier, A.; Madore, J.; O’Donnell, J.S.; Johnston, R.L.; Li, X.Y.; McDonald, E.; Ahern, E.; Kuchel, A.; Eastgate, M.; Pearson, S.-A.; et al. Tumor CD155 expression is associated with resistance to anti-PD1 immunotherapy in metastatic melanoma. Clin. Cancer Res. 2020, 26, 3671–3681. [Google Scholar] [CrossRef]
  100. Sun, Y.; Luo, J.; Chen, Y.; Cui, J.; Lei, Y.; Cui, Y.; Jiang, N.; Jiang, W.; Chen, L.; Chen, 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. [Google Scholar] [CrossRef]
  101. Wen, J.; Mao, X.; Cheng, Q.; Liu, Z.; Liu, F. A pan-cancer analysis revealing the role of TIGIT in tumor microenvironment. Sci. Rep. 2021, 11, 22502. [Google Scholar] [CrossRef] [PubMed]
  102. Waight, J.D.; Chand, D.; Dietrich, S.; Gombos, R.; Horn, T.; Gonzalez, A.M.; Manrique, M.; Swiech, L.; Morin, B.; Brittsan, C.; et al. Selective FcγR Co-engagement on APCs modulates the activity of therapeutic antibodies targeting T cell antigens. Cancer Cell 2018, 33, 1033–1047.e5. [Google Scholar] [CrossRef]
  103. Sivick Gauthier, K.E.; Piovesan, D.; Groot, A.E.D.; Anderson, A.E.; Ray, R.D.; Serwas, N.K.; Cho, S.; Patnaik, A.; Foster, P.; Mitchell, C.G.; et al. 475 Fc-Silent Anti-TIGIT Antibodies Potentiate Anti-Tumor Immunity without Depleting Regulatory T Cells. J. ImmunoTherapy Cancer 2023, 11, A534. [Google Scholar] [CrossRef]
  104. Cho, B.C.; Abreu, D.R.; Hussein, M.; Cobo, M.; Patel, A.J.; Secen, N.; Lee, K.H.; Massuti, B.; Hiret, S.; Yang, J.C.H.; 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, 781–792. [Google Scholar] [CrossRef]
  105. Guan, X.; Hu, R.; Choi, Y.; Srivats, S.; Nabet, B.Y.; Silva, J.; McGinnis, L.; Hendricks, R.; Nutsch, K.; Banta, K.L.; et al. Anti-TIGIT antibody improves PD-L1 blockade through myeloid and treg cells. Nature 2024, 627, 646–655. [Google Scholar] [CrossRef] [PubMed]
  106. Roche Reports Update on Phase III SKYSCRAPER-01 Study Results. Available online: https://www.roche.com/media/releases/med-cor-2024-11-26 (accessed on 26 November 2024).
  107. Socinski, M.A.; Rodriguez Abreu, D.; Lee, D.H.; Cappuzzo, F.; Nishio, M.; Lovly, C.M.; Ozyilkan, O.; Li, Q.; Johnson, M.L.; Garon, E.B.; et al. LBA2 SKYSCRAPER-06: Efficacy and safety of tiragolumab plus atezolizumab plus chemotherapy (Tira + Atezo + Chemo) vs pembrolizumab plus chemotherapy (Pembro + Chemo) in patients (Pts) with advanced non-squamous non-small cell lung cancer (NSq NSCLC). Immuno-Oncol. Technol. 2024, 24, 101025. [Google Scholar] [CrossRef]
  108. Johnson, M.L.; Fox, W.; Lee, Y.G.; Lee, K.H.; Ahn, H.K.; Kim, Y.C.; Lee, K.Y.; Lee, J.S.; He, X.; Park, C.; et al. ARC-7: Randomized phase 2 study of domvanalimab + zimberelimab ± etrumadenant versus zimberelimab in first-line, metastatic, PD-L1-high non-small cell lung cancer (NSCLC). J. Clin. Oncol. 2022, 40, 397600. [Google Scholar] [CrossRef]
  109. Naidoo, J.; Peters, S.; Runglodvatana, Y.; Li, J.Y.C.; Fong, C.H.; Ho, G.F.; How, S.H.; Juengsamarn, J.; Todd, T.; Marina, N.; et al. 1461 phase 2 randomized study of domvanalimab combined with zimberelimab in front-line, PD-(L)1 high, locally advanced or metastatic non-small cell lung cancer (NSCLC): Results from ARC-10 part 1. J. ImmunoTherapy Cancer 2024, 12. [Google Scholar] [CrossRef]
  110. Arcus Biosciences Announces That Domvanalimab Plus Zimberelimab Improved Overall Survival in ARC-10, a Randomized Study in Patients with PD-L1-High Non-Small Cell Lung Cancer. Available online: https://investors.arcusbio.com/investors-and-media/press-releases/press-release-details/2024/Arcus-Biosciences-Announces-that-Domvanalimab-Plus-Zimberelimab-Improved-Overall-Survival-in-ARC-10-a-Randomized-Study-in-Patients-with-PD-L1-High-Non-Small-Cell-Lung-Cancer/default.aspx (accessed on 5 November 2024).
  111. Rodriguez-Abreu, D.; Gray, J.E.; Ahn, M.J.; Johnson, M.L.; Yu, X.; Chen, X.; Phuong, P.H.; Kim, J.; Reck, M. STAR-121: A phase 3, randomized study of Domvanalimab (DOM) and Zimberelimab (ZIM) in combination with chemotherapy vs pembrolizumab (Pembro) and chemotherapy in patients with untreated metastatic non-small cell lung cancer (mNSCLC) with no actionable gene alterations. J. Clin. Oncol. 2023, 41, TPS9141. [Google Scholar] [CrossRef]
  112. Spira, A.I.; Chiu, J.; Wang, C.C.; Zer, A.; Conibear, J.; Phuong, P.H.; Park, J.K.; Seto, A.; Zhang, J.; Cho, B.C. VELOCITY-lung: A phase (Ph) 2 study evaluating safety and efficacy of domvanalimab (Dom) + Zimberelimab (Zim) + Sacituzumab Govitecan (SG), or Etrumadenant (Etruma) + Dom + Zim, or Etruma + Zim in patients (Pts) with treatment-naïve metastatic non-small cell lung cancer (mNSCLC). J. Clin. Oncol. 2023, 41, TPS9155. [Google Scholar] [CrossRef]
  113. Niu, J.; Maurice-Dror, C.; Lee, D.H.; Kim, D.W.; Nagrial, A.; Voskoboynik, M.; Chung, H.C.; Mileham, K.; Vaishampayan, U.; Rasco, D.; 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, 169–180. [Google Scholar] [CrossRef] [PubMed]
  114. Peled, N.; Mazieres, J.; Kowalski, D.M.; Lam, W.S.; Hochmair, M.J.; Majem, M.; Kim, S.H.; Blanco, A.C.; Cuppens, K.; Casarini, I.A.; et al. 121P MK-7684A (Vibostolimab [Vibo] plus Pembrolizumab [Pembro] coformulation) with/without Docetaxel in metastatic NSCLC after platinum-chemotherapy (Chemo) and immunotherapy. Immuno. Oncol. Technol. 2023, 20, 100593. [Google Scholar] [CrossRef]
  115. Cho, B.C.; Juergens, R.A.; Cheng, Y.; De Castro, G.; Erman, M.; Bauman, J.R.; Takahashi, T.; Schwarzenberger, P.; Li, C.; Pietanza, M.C.; et al. Abstract CT561: KeyVibe-003: Randomized, double-blind, phase 3 study of first-line pembrolizumab with and without vibostolimab (Anti-TIGIT) in patients with PD-L1-positive metastatic NSCLC. Cancer Res. 2022, 82, CT561. [Google Scholar] [CrossRef]
  116. Garassino, M.C.; Felip, E.; Awad, M.M.; Wang, W.; Kim, S.J.; Pietanza, M.C.; Rodriguez-Abreu, D. A randomized, double-blind, phase 3 trial of MK-7684A plus chemotherapy versus pembrolizumab plus chemotherapy as first-line therapy for metastatic non–small cell lung cancer (NSCLC): KeyVibe-007. J. Clin. Oncol. 2022, 40, TPS9160. [Google Scholar] [CrossRef]
  117. Rohrberg, K.S.; Brandão, M.; Castanon Alvarez, E.; Felip, E.; Gort, E.H.; Hiltermann, T.J.J.N.; Izumi, H.; Kim, D.W.; Kim, S.W.; Paz-Ares, L.G.; et al. Safety, Pharmacokinetics (PK), Pharmacodynamics (PD) and preliminary efficacy of AZD2936, a bispecific antibody targeting PD-1 and TIGIT, in checkpoint inhibitor (CPI)-experienced advanced/metastatic non-small-cell lung cancer (NSCLC): First report of ARTEMIDE-01. J. Clin. Oncol. 2023, 41, 9050. [Google Scholar] [CrossRef]
  118. Brandão, M.; Hiltermann, J.T.J.N.; Wauters, E.; Solomon, B.J.; Castanon Alvarez, E.; Felip, E.; Gort, E.H.; Izumi, H.; Kim, D.W.; Paz-Ares, L.; et al. 1446P preliminary efficacy and safety of rilvegostomig (AZD2936), a bispecific antibody targeting PD-1 and TIGIT, in checkpoint inhibitor (CPI)-pretreated advanced/metastatic non-small-cell lung cancer (NSCLC): ARTEMIDE-01. Ann. Oncol. 2023, 34, S822–S823. [Google Scholar] [CrossRef]
  119. Hiltermann, T.J.N.; Izumi, H.; Cho, B.C.; Cunha, S.; Danchaivijitr, P.; Felip, E.; Ho, G.F.; Leventakos, K.; Li, Y.; Sugawara, S.; et al. OA11.03 efficacy and safety of Rilvegostomig, an anti-PD-1/TIGIT bispecific, for CPI-naïve metastatic NSCLC with PD-L1 1-49% or ≥50%. J. Thorac. Oncol. 2024, 19, S33. [Google Scholar] [CrossRef]
  120. Baixeras, E.; Huard, B.; Miossec, C.; Jitsukawa, S.; Martin, M.; Hercend, T.; Auffray, C.; Triebel, F.; Piatier-Tonneau, D. Characterization of the lymphocyte activation gene 3-encoded protein. A new ligand for human leukocyte antigen class II antigens. J. Exp. Med. 1992, 176, 327–337. [Google Scholar] [CrossRef]
  121. Andrews, L.P.; Yano, H.; Vignali, D.A.A. Inhibitory receptors and ligands beyond PD-1, PD-L1 and CTLA-4: Breakthroughs or backups. Nat. Immunol. 2019, 20, 1425–1434. [Google Scholar] [CrossRef]
  122. Triebel, F.; Jitsukawa, S.; Baixeras, E.; Roman-Roman, S.; Genevee, C.; Viegas-Pequignot, E.; Hercend, T. LAG-3, a novel lymphocyte activation gene closely related to CD4. J. Exp. Med. 1990, 171, 1393–1405. [Google Scholar] [CrossRef]
  123. Guy, C.; Mitrea, D.M.; Chou, P.C.; Temirov, J.; Vignali, K.M.; Liu, X.; Zhang, H.; Kriwacki, R.; Bruchez, M.P.; Watkins, S.C.; et al. LAG3 associates with TCR–CD3 complexes and suppresses signaling by driving co-receptor–Lck dissociation. Nat. Immunol. 2022, 23, 757–767. [Google Scholar] [CrossRef] [PubMed]
  124. Wang, J.; Sanmamed, M.F.; Datar, I.; Su, T.T.; Ji, L.; Sun, J.; Chen, L.; Chen, Y.; Zhu, G.; Yin, W.; et al. Fibrinogen-like protein 1 is a major immune inhibitory ligand of LAG-3. Cell 2019, 176, 334–347.e12. [Google Scholar] [CrossRef]
  125. Kouo, T.; Huang, L.; Pucsek, A.B.; Cao, M.; Solt, S.; Armstrong, T.; Jaffee, E. Galectin-3 shapes antitumor immune responses by suppressing CD8+ T cells via LAG-3 and inhibiting expansion of plasmacytoid dendritic cells. Cancer Immunol. Res. 2015, 3, 412–423. [Google Scholar] [CrossRef] [PubMed]
  126. Xu, F.; Liu, J.; Liu, D.; Liu, B.; Wang, M.; Hu, Z.; Du, X.; Tang, L.; He, F. LSECtin expressed on melanoma cells promotes tumor progression by inhibiting antitumor T-Cell responses. Cancer Res. 2014, 74, 3418–3428. [Google Scholar] [CrossRef] [PubMed]
  127. Triebel, F. LAG-3: A regulator of T-cell and DC responses and its use in therapeutic vaccination. Trends Immunol. 2003, 24, 619–622. [Google Scholar] [CrossRef]
  128. Andrews, L.P.; Somasundaram, A.; Moskovitz, J.M.; Szymczak-Workman, A.L.; Liu, C.; Cillo, A.R.; Lin, H.; Normolle, D.P.; Moynihan, K.D.; Taniuchi, I.; et al. Resistance to PD1 blockade in the absence of metalloprotease-mediated LAG3 shedding. Sci. Immunol. 2020, 5, eabc2728. [Google Scholar] [CrossRef]
  129. Liang, B.; Workman, C.; Lee, J.; Chew, C.; Dale, B.M.; Colonna, L.; Flores, M.; Li, N.; Schweighoffer, E.; Greenberg, S.; et al. Regulatory T cells inhibit dendritic cells by lymphocyte activation gene-3 engagement of MHC class II. J. Immunol. 2008, 180, 5916–5926. [Google Scholar] [CrossRef]
  130. Huang, C.T.; Workman, C.J.; Flies, D.; Pan, X.; Marson, A.L.; Zhou, G.; Hipkiss, E.L.; Ravi, S.; Kowalski, J.; Levitsky, H.I.; et al. Role of LAG-3 in regulatory T cells. Immunity 2004, 21, 503–513. [Google Scholar] [CrossRef]
  131. Zhang, Q.; Beres, A.; Vignali, D. Regulatory T cells use LAG-3 as the “brake” in autoimmune diabetes but the “accelerator” in tumor environment (IRC4P.487). J. Immunol. 2014, 192, 60.14. [Google Scholar] [CrossRef]
  132. Thaker, Y.R.; Andrews, L.P.; Workman, C.J.; Vignali, D.A.A.; Sharpe, A.H. Treg-specific LAG3 deletion reveals a key role for LAG3 in regulatory T Cells to inhibit CNS autoimmunity. J. Immunol. 2018, 200 (Suppl. S1), 101.7. [Google Scholar] [CrossRef]
  133. Kim, D.; Kim, G.; Yu, R.; Lee, J.; Kim, S.; Gleason, M.R.; Qiu, K.; Montauti, E.; Wang, L.L.; Fang, D.; et al. Inhibitory co-receptor Lag3 supports Foxp3+ regulatory T cell function by restraining Myc-dependent metabolic programming. Immunity 2024, 57, 2634–2650.e5. [Google Scholar] [CrossRef]
  134. Dykema, A.G.; Zhang, J.; Cheung, L.S.; Connor, S.; Zhang, B.; Zeng, Z.; Cherry, C.M.; Li, T.; Caushi, J.X.; Nishimoto, M.; et al. Lung tumor–infiltrating treg have divergent transcriptional profiles and function linked to checkpoint blockade response. Sci. Immunol. 2023, 8, eadg1487. [Google Scholar] [CrossRef]
  135. Su, J.; Fu, Y.; Cui, Z.; Abidin, Z.; Yuan, J.; Zhang, X.; Li, R.; Zhao, C. Relatlimab: A novel drug targeting immune checkpoint LAG-3 in melanoma therapy. Front. Pharmacol. 2024, 14, 1349081. [Google Scholar] [CrossRef] [PubMed]
  136. Detterbeck, F.C. The eighth edition TNM stage classification for lung cancer: What does it mean on main street? J. Thorac. Cardiovasc. Surg. 2018, 155, 356–359. [Google Scholar] [CrossRef] [PubMed]
  137. Girard, N.; Burotto, M.; Paz-Ares, L.G.; Reck, M.; Schenker, M.; Lingua, A.; Orlandi, F.J.; Naidoo, J.; Beardlsey, E.K.; Velcheti, V.; et al. LBA53 nivolumab (NIVO) plus relatlimab with platinum-doublet chemotherapy (PDCT) vs NIVO + PDCT as first-line (1L) treatment (Tx) for stage IV or recurrent NSCLC: Results from the randomized phase II RELATIVITY-104 study. Ann. Oncol. 2024, 35, S1243–S1244. [Google Scholar] [CrossRef]
  138. Schöffski, P.; Tan, D.S.W.; Martín, M.; Ochoa-de-Olza, M.; Sarantopoulos, J.; Carvajal, R.D.; Kyi, C.; Esaki, T.; Prawira, A.; Akerley, W.; et al. phase I/II study of the LAG-3 inhibitor ieramilimab (LAG525) ± anti-PD-1 spartalizumab (PDR001) in patients with advanced malignancies. J. Immunother. Cancer. 2022, 10, e003776. [Google Scholar] [CrossRef]
  139. Lin, C.C.; Garralda, E.; Schöffski, P.; Hong, D.S.; Siu, L.L.; Martin, M.; Maur, M.; Hui, R.; Soo, R.A.; Chiu, J.; et al. A phase 2, multicenter, open-label study of anti-LAG-3 ieramilimab in combination with anti-PD-1 spartalizumab in patients with advanced solid malignancies. OncoImmunology 2024, 13, 2290787. [Google Scholar] [CrossRef]
  140. Mishra, A.K.; Shahid, S.; Karade, S.S.; Agnihotri, P.; Kolesnikov, A.; Hasan, S.S.; Mariuzza, R.A. CryoEM structure of a therapeutic antibody (Favezelimab) bound to human LAG3 determined using a bivalent fab as fiducial marker. Structure 2023, 31, 1149–1157.e3. [Google Scholar] [CrossRef]
  141. Gutierrez, M.; Lam, W.S.; Hellmann, M.; Gubens, M.; Aggarwal, C.; Weng Tan, D.S.; Felip, E.; Chiu, J.W.; Lee, J.S.; Yang, J.C.-H.; et al. 457 KEYNOTE-495/KeyImPaCT: Interim Analysis of a Randomized, Biomarker-Directed, Phase 2 Trial of Pembrolizumab-Based Combination Therapy for Non–Small Cell Lung Cancer (NSCLC). J. ImmunoTherapy Cancer 2021, 9. [Google Scholar] [CrossRef]
  142. Gutierrez, M.; Lam, W.S.; Hellmann, M.D.; Gubens, M.A.; Aggarwal, C.; Tan, D.S.W.; Felip, E.; Chiu, J.W.Y.; Lee, J.S.; Yang, J.C.H.; et al. Biomarker-directed, pembrolizumab-based combination therapy in non-small cell lung cancer: Phase 2 KEYNOTE-495/KeyImPaCT trial interim results. Nat. Med. 2023, 29, 1718–1727. [Google Scholar] [CrossRef]
  143. Faulkner, N.E.; Melkadze, T.; Nair, S.; Gabrail, N.; Kunta, G.; Ibrahim, E.; Dreisbach, L.; Brungs, D.; Chikhladze, N.; Gogishvili, M.; et al. A phase 2/3 study of fianlimab plus cemiplimab versus cemiplimab in patients with advanced non-small cell lung cancer with tumors expressing PD-L1 ≥50%. J. Clin. Oncol. 2024, 42, TPS8663. [Google Scholar] [CrossRef]
  144. Gabrail, N.; Gogishvili, M.; Makharadze, T.; Chikhladze, N.; Faulkner, N.E.; Nair, S.; Ibrahim, E.; Brungs, D.; Dreisbach, L.; Kunta, G.; et al. A phase 2/3 study of fianlimab, cemiplimab, plus chemotherapy versus cemiplimab plus chemotherapy in first-line treatment of advanced non-small cell lung cancer. J. Clin. Oncol. 2024, 42, TPS8660. [Google Scholar] [CrossRef]
  145. Johnson, M.L.; Patel, M.R.; Ulahannan, S.V.; Hansen, A.; George, B.; Chu, Q.S.C.; Elgadi, M.; Ge, M.; Duffy, C.; Graeser, R.; et al. Phase I study of BI 754111 (Anti-LAG-3) plus BI 754091(Anti-PD-1) in patients (Pts) with advanced solid cancers, followed by expansion in pts with microsatellite stable metastatic colorectal cancer (mCRC), anti-PD-(L)1-pretreated non-small cell lung cancer (NSCLC) and other solid tumors. Ann. Oncol. 2018, 29, viii441. [Google Scholar] [CrossRef]
  146. Johnson, M.L.; Patel, M.R.; Cherry, M.; Kang, Y.K.; Yamaguchi, K.; Oh, D.Y.; Hussein, M.A.; Kitano, S.; Kondo, S.; Hansen, A.R.; et al. Safety of BI 754111, an anti-LAG-3 monoclonal antibody (mAb), in combination with BI 754091, an anti-PD-1 mAb, in patients with advanced solid tumors. J. Clin. Oncol. 2020, 38, 3063. [Google Scholar] [CrossRef]
  147. Zettl, M.; Wurm, M.; Schaaf, O.; Mostböck, S.; Tirapu, I.; Apfler, I.; Lorenz, I.C.; Frego, L.; Kenny, C.; Thibodeau, M.; et al. Combination of two novel blocking antibodies, anti-PD-1 antibody ezabenlimab (BI 754091) and anti-LAG-3 antibody BI 754111, leads to increased immune cell responses. OncoImmunology 2022, 11, 2080328. [Google Scholar] [CrossRef]
  148. Ghosh, S.; Sharma, G.; Travers, J.; Kumar, S.; Choi, J.; Jun, H.T.; Kehry, M.; Ramaswamy, S.; Jenkins, D. TSR-033, a novel therapeutic antibody targeting LAG-3, enhances T-cell function and the activity of PD-1 blockade in vitro and in vivo. Mol. Cancer Ther. 2019, 18, 632–641. [Google Scholar] [CrossRef]
  149. Grandal, M.M.; Melander, M.C.; Bhatia, V.K.; Gjetting, T.; Lindsted, T.; Fröhlich, C.; Lantto, J.; Horak, I.D.; Kragh, M.; Kofoed, K.; et al. Abstract 5626: Preclinical characterization of Sym022, a novel anti-LAG3 antibody. Cancer Res. 2018, 78, 5626. [Google Scholar] [CrossRef]
  150. Yap, T.A.; LoRusso, P.M.; Wong, D.J.; Hu-Lieskovan, S.; Papadopoulos, K.P.; Holz, J.B.; Grabowska, U.; Gradinaru, C.; Leung, K.M.; Marshall, S.; et al. A phase 1 first-in-human study of FS118, a tetravalent bispecific antibody targeting LAG-3 and PD-L1 in patients with advanced cancer and PD-L1 resistance. Clin. Cancer Res. 2023, 29, 888–898. [Google Scholar] [CrossRef]
  151. Rohrberg, K.S.; Garralda, E.; Calvo, E.; Moreno Garcia, V.; Guidi, M.; Kraus, D.G.; McIntyre, C.; Kao, H.; Codarri Deak, L.; Michielin, F.; et al. 745P clinical activity, safety, and PK/PD from the first in human study (NP41300) of RO7247669, a PD1-LAG3 bispecific antibody. Ann. Oncol. 2022, 33, S884–S885. [Google Scholar] [CrossRef]
  152. Luke, J.J.; Patel, M.R.; Blumenschein, G.R.; Hamilton, E.; Chmielowski, B.; Ulahannan, S.V.; Connolly, R.M.; Santa-Maria, C.A.; Wang, J.; Bahadur, S.W.; et al. The PD-1- and LAG-3-targeting bispecific molecule tebotelimab in solid tumors and hematologic cancers: A phase 1 trial. Nat. Med. 2023, 29, 2814–2824. [Google Scholar] [CrossRef]
  153. Ma, J.; Mo, Y.; Tang, M.; Shen, J.; Qi, Y.; Zhao, W.; Huang, Y.; Xu, Y.; Qian, C. Bispecific antibodies: From research to clinical application. Front. Immunol. 2021, 12, 626616. [Google Scholar] [CrossRef] [PubMed]
  154. Acharya, N.; Sabatos-Peyton, C.; Anderson, A.C. Tim-3 finds its place in the cancer immunotherapy landscape. J. Immunother. Cancer 2020, 8, e000911. [Google Scholar] [CrossRef]
  155. Wolf, Y.; Anderson, A.C.; Kuchroo, V.K. TIM3 comes of age as an inhibitory receptor. Nat. Rev. Immunol. 2020, 20, 173–185. [Google Scholar] [CrossRef]
  156. Dixon, K.O.; Lahore, G.F.; Kuchroo, V.K. Beyond T cell exhaustion: TIM-3 regulation of myeloid cells. Sci. Immunol. 2024, 9, eadf2223. [Google Scholar] [CrossRef] [PubMed]
  157. Monney, L.; Sabatos, C.A.; Gaglia, J.L.; Ryu, A.; Waldner, H.; Chernova, T.; Manning, S.; Greenfield, E.A.; Coyle, A.J.; Sobel, R.A.; et al. Th1-specific cell surface protein Tim-3 regulates macrophage activation and severity of an autoimmune disease. Nature 2002, 415, 536–541. [Google Scholar] [CrossRef]
  158. Ndhlovu, L.C.; Lopez-Vergès, S.; Barbour, J.D.; Jones, R.B.; Jha, A.R.; Long, B.R.; Schoeffler, E.C.; Fujita, T.; Nixon, D.F.; Lanier, L.L. Tim-3 marks human natural killer cell maturation and suppresses cell-mediated cytotoxicity. Blood 2012, 119, 3734–3743. [Google Scholar] [CrossRef]
  159. Gao, X.; Zhu, Y.; Li, G.; Huang, H.; Zhang, G.; Wang, F.; Sun, J.; Yang, Q.; Zhang, X.; Lu, B. TIM-3 expression characterizes regulatory t cells in tumor tissues and is associated with lung cancer progression. PLoS ONE 2012, 7, e30676. [Google Scholar] [CrossRef] [PubMed]
  160. Komohara, Y.; Morita, T.; Annan, D.A.; Horlad, H.; Ohnishi, K.; Yamada, S.; Nakayama, T.; Kitada, S.; Suzu, S.; Kinoshita, I.; et al. The coordinated actions of TIM-3 on cancer and myeloid cells in the regulation of tumorigenicity and clinical prognosis in clear cell renal cell carcinomas. Cancer Immunol. Res. 2015, 3, 999–1007. [Google Scholar] [CrossRef]
  161. Dixon, K.O.; Tabaka, M.; Schramm, M.A.; Xiao, S.; Tang, R.; Dionne, D.; Anderson, A.C.; Rozenblatt-Rosen, O.; Regev, A.; Kuchroo, V.K. TIM-3 restrains anti-tumour immunity by regulating inflammasome activation. Nature 2021, 595, 101–106. [Google Scholar] [CrossRef]
  162. Phong, B.L.; Avery, L.; Sumpter, T.L.; Gorman, J.V.; Watkins, S.C.; Colgan, J.D.; Kane, L.P. Tim-3 enhances FcεRI-proximal signaling to modulate mast cell activation. J. Exp. Med. 2015, 212, 2289–2304. [Google Scholar] [CrossRef]
  163. Zhu, C.; Anderson, A.C.; Schubart, A.; Xiong, H.; Imitola, J.; Khoury, S.J.; Zheng, X.X.; Strom, T.B.; Kuchroo, V.K. The Tim-3 ligand Galectin-9 negatively regulates t helper type 1 immunity. Nat. Immunol. 2005, 6, 1245–1252. [Google Scholar] [CrossRef] [PubMed]
  164. Lv, Y.; Ma, X.; Ma, Y.; Du, Y.; Feng, J. A New emerging target in cancer immunotherapy: Galectin-9 (LGALS9). Genes Dis. 2023, 10, 2366–2382. [Google Scholar] [CrossRef]
  165. Elahi, S.; Niki, T.; Hirashima, M.; Horton, H. Galectin-9 binding to Tim-3 renders activated human CD4+ T cells less susceptible to HIV-1 infection. Blood 2012, 119, 4192–4204. [Google Scholar] [CrossRef] [PubMed]
  166. Rahmati, A.; Bigam, S.; Elahi, S. Galectin-9 promotes natural killer cells activity via interaction with CD44. Front. Immunol. 2023, 14, 1131379. [Google Scholar] [CrossRef]
  167. Huang, Y.H.; Zhu, C.; Kondo, Y.; Anderson, A.C.; Gandhi, A.; Russell, A.; Dougan, S.K.; Petersen, B.S.; Melum, E.; Pertel, T.; et al. CEACAM1 regulates TIM-3-mediated tolerance and exhaustion. Nature 2015, 517, 386–390. [Google Scholar] [CrossRef]
  168. Huang, Y.H.; Yoon, C.H.; Gandhi, A.; Hanley, T.; Castrillon, C.; Kondo, Y.; Lin, X.; Kim, W.; Yang, C.; Driouchi, A.; et al. High-dimensional mapping of human CEACAM1 expression on immune cells and association with melanoma drug resistance. Commun. Med. 2024, 4, 128. [Google Scholar] [CrossRef]
  169. Takeuchi, A.; Yokoyama, S.; Nakamori, M.; Nakamura, M.; Ojima, T.; Yamaguchi, S.; Mitani, Y.; Shively, J.E.; Yamaue, H. Loss of CEACAM1 is associated with poor prognosis and peritoneal dissemination of patients with gastric cancer. Sci. Rep. 2019, 9, 12702. [Google Scholar] [CrossRef]
  170. DeKruyff, R.H.; Bu, X.; Ballesteros, A.; Santiago, C.; Chim, Y.L.E.; Lee, H.H.; Karisola, P.; Pichavant, M.; Kaplan, G.G.; Umetsu, D.T.; et al. T Cell/transmembrane, Ig, and Mucin-3 allelic variants differentially recognize phosphatidylserine and mediate phagocytosis of apoptotic cells. J. Immunol. 2010, 184, 1918–1930. [Google Scholar] [CrossRef] [PubMed]
  171. Chiba, S.; Baghdadi, M.; Akiba, H.; Yoshiyama, H.; Kinoshita, I.; Dosaka-Akita, H.; Fujioka, Y.; Ohba, Y.; Gorman, J.V.; Colgan, J.D.; et al. Tumor-infiltrating DCs suppress nucleic acid–mediated innate immune responses through interactions between the receptor TIM-3 and the alarmin HMGB1. Nat. Immunol. 2012, 13, 832–842. [Google Scholar] [CrossRef]
  172. Rangachari, M.; Zhu, C.; Sakuishi, K.; Xiao, S.; Karman, J.; Chen, A.; Angin, M.; Wakeham, A.; Greenfield, E.A.; Sobel, R.A.; et al. Bat3 promotes T cell responses and autoimmunity by repressing Tim-3–mediated cell death and exhaustion. Nat. Med. 2012, 18, 1394–1400. [Google Scholar] [CrossRef]
  173. Van De Weyer, P.S.; Muehlfeit, M.; Klose, C.; Bonventre, J.V.; Walz, G.; Kuehn, E.W. A highly conserved tyrosine of Tim-3 is phosphorylated upon stimulation by its ligand Galectin-9. Biochem. Biophys. Res. Commun. 2006, 351, 571–576. [Google Scholar] [CrossRef] [PubMed]
  174. Davidson, D.; Schraven, B.; Veillette, A. PAG-associated fynt regulates calcium signaling and promotes anergy in T lymphocytes. Mol. Cell. Biol. 2007, 27, 1960–1973. [Google Scholar] [CrossRef] [PubMed]
  175. Lee, J.; Su, E.W.; Zhu, C.; Hainline, S.; Phuah, J.; Moroco, J.A.; Smithgall, T.E.; Kuchroo, V.K.; Kane, L.P. Phosphotyrosine-dependent coupling of Tim-3 to T-cell receptor signaling pathways. Mol. Cell. Biol. 2011, 31, 3963–3974. [Google Scholar] [CrossRef]
  176. Clayton, K.L.; Haaland, M.S.; Douglas-Vail, M.B.; Mujib, S.; Chew, G.M.; Ndhlovu, L.C.; Ostrowski, M.A. T cell Ig and mucin domain–containing protein 3 is recruited to the immune synapse, disrupts stable synapse formation, and associates with receptor phosphatases. J. Immunol. 2014, 192, 782–791. [Google Scholar] [CrossRef]
  177. Bertheloot, D.; Latz, E. HMGB1, IL-1α, IL-33 and S100 proteins: Dual-function alarmins. Cell Mol. Immunol. 2017, 14, 43–64. [Google Scholar] [CrossRef] [PubMed]
  178. De Mingo Pulido, Á.; Hänggi, K.; Celias, D.P.; Gardner, A.; Li, J.; Batista-Bittencourt, B.; Mohamed, E.; Trillo-Tinoco, J.; Osunmakinde, O.; Peña, R.; et al. The inhibitory receptor TIM-3 limits activation of the cGAS-STING pathway in intra-tumoral dendritic cells by suppressing extracellular DNA uptake. Immunity 2021, 54, 1154–1167.e7. [Google Scholar] [CrossRef]
  179. Redmond, W.L.; Sherman, L.A. Peripheral tolerance of CD8 T lymphocytes. Immunity 2005, 22, 275–284. [Google Scholar] [CrossRef]
  180. Luckashenak, N.; Schroeder, S.; Endt, K.; Schmidt, D.; Mahnke, K.; Bachmann, M.F.; Marconi, P.; Deeg, C.A.; Brocker, T. Constitutive crosspresentation of tissue antigens by dendritic cells controls CD8+ T cell tolerance in vivo. Immunity 2008, 28, 521–532. [Google Scholar] [CrossRef]
  181. Nakayama, M.; Akiba, H.; Takeda, K.; Kojima, Y.; Hashiguchi, M.; Azuma, M.; Yagita, H.; Okumura, K. Tim-3 mediates phagocytosis of apoptotic cells and cross-presentation. Blood 2009, 113, 3821–3830. [Google Scholar] [CrossRef]
  182. Yan, J.; Zhang, Y.; Zhang, J.P.; Liang, J.; Li, L.; Zheng, L. Tim-3 expression defines regulatory T cells in human tumors. PLoS ONE 2013, 8, e58006. [Google Scholar] [CrossRef]
  183. Banerjee, H.; Nieves-Rosado, H.; Kulkarni, A.; Murter, B.; McGrath, K.V.; Chandran, U.R.; Chang, A.; Szymczak-Workman, A.L.; Vujanovic, L.; Delgoffe, G.M.; et al. Expression of Tim-3 drives phenotypic and functional changes in treg cells in secondary lymphoid organs and the tumor microenvironment. Cell Rep. 2021, 36, 109699. [Google Scholar] [CrossRef]
  184. Sakuishi, K.; Ngiow, S.F.; Sullivan, J.M.; Teng, M.W.L.; Kuchroo, V.K.; Smyth, M.J.; Anderson, A.C. TIM3+ FOXP3+ regulatory T cells are tissue-specific promoters of T-cell dysfunction in cancer. OncoImmunology 2013, 2, e23849. [Google Scholar] [CrossRef]
  185. Zhu, C.; Dixon, K.O.; Newcomer, K.; Gu, G.; Xiao, S.; Zaghouani, S.; Schramm, M.A.; Wang, C.; Zhang, H.; Goto, K.; et al. Tim-3 adaptor protein Bat3 is a molecular checkpoint of T cell terminal differentiation and exhaustion. Sci. Adv. 2021, 7, eabd2710. [Google Scholar] [CrossRef] [PubMed]
  186. Jansen, C.S.; Prokhnevska, N.; Master, V.A.; Sanda, M.G.; Carlisle, J.W.; Bilen, M.A.; Cardenas, M.; Wilkinson, S.; Lake, R.; Sowalsky, A.G.; et al. An intra-tumoral niche maintains and differentiates stem-like CD8 T cells. Nature 2019, 576, 465–470. [Google Scholar] [CrossRef] [PubMed]
  187. Beltra, J.C.; Manne, S.; Abdel-Hakeem, M.S.; Kurachi, M.; Giles, J.R.; Chen, Z.; Casella, V.; Ngiow, S.F.; Khan, O.; Huang, Y.J.; et al. Developmental relationships of four exhausted CD8+ T cell subsets reveals underlying transcriptional and epigenetic landscape control mechanisms. Immunity 2020, 52, 825–841.e8. [Google Scholar] [CrossRef]
  188. Koyama, S.; Akbay, E.A.; Li, Y.Y.; Herter-Sprie, G.S.; Buczkowski, K.A.; Richards, W.G.; Gandhi, L.; Redig, A.J.; Rodig, S.J.; Asahina, H.; et al. Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat. Commun. 2016, 7, 10501. [Google Scholar] [CrossRef]
  189. Falchook, G.S.; Ribas, A.; Davar, D.; Eroglu, Z.; Wang, J.S.; Luke, J.J.; Hamilton, E.P.; Di Pace, B.; Wang, T.; Ghosh, S.; et al. Phase 1 trial of TIM-3 inhibitor cobolimab monotherapy and in combination with PD-1 inhibitors nivolumab or dostarlimab (AMBER). J. Clin. Oncol. 2022, 40, 2504. [Google Scholar] [CrossRef]
  190. Davar, D.; Eroglu, Z.; Milhem, M.; Becerra, C.; Gutierrez, M.; Ribas, A.; Pace, B.D.; Wang, T.; Zhang, H.; Ghosh, S.; et al. 596 AMBER, part 2B: A phase 1 study of cobolimab plus dostarlimab in patients with advanced/metastatic non-small cell lung cancer (NSCLC) previously treated with anti-PD(L)-1 therapy. J. ImmunoTherapy Cancer 2023, 11. [Google Scholar] [CrossRef]
  191. Kim, H.R.; Gridelli, C.; Kapur, D.; Tufman, A.; Felip, E.; Velcheti, V.; Kim, Y.J.; Goetze, T.O.; Lopez, P.G.; Corre, R.; et al. P1.11-01 cobolimab with dostarlimab and docetaxel in patients with advanced non-small cell lung cancer (NSCLC): COSTAR lung. J. Thorac. Oncol. 2022, 17, S109–S110. [Google Scholar] [CrossRef]
  192. Mach, N.; Curigliano, G.; Santoro, A.; Kim, D.W.; Tai, D.W.M.; Hodi, S.; Wilgenhof, S.; Doi, T.; Longmire, T.; Sun, H.; et al. Phase (Ph) II study of MBG453 + Spartalizumab in patients (Pts) with non-small cell lung cancer (NSCLC) and melanoma pretreated with Anti–PD-1/L1 therapy. Ann. Oncol. 2019, 30, v491–v492. [Google Scholar] [CrossRef]
  193. Harding, J.J.; Moreno, V.; Bang, Y.J.; Hong, M.H.; Patnaik, A.; Trigo, J.; Szpurka, A.M.; Yamamoto, N.; Doi, T.; Fu, S.; et al. Blocking TIM-3 in treatment-refractory advanced solid tumors: A phase Ia/b Study of LY3321367 with or without an anti-PD-L1 antibody. Clin. Cancer Res. 2021, 27, 2168–2178. [Google Scholar] [CrossRef]
  194. Waight, J.; Iyer, P.; Breous-Nystrom, E.; Riordan, C.; Findeis, M.; Underwood, D.; Connolly, J.; Sanicola-Nadel, M.; Nastri, H.; Scherle, P.; et al. Abstract 3825: INCAGN02390, a novel antagonist antibody that targets the co-inhibitory receptor TIM-3. Cancer Res. 2018, 78, 3825. [Google Scholar] [CrossRef]
  195. Besse, B.; Italiano, A.; Cousin, S.; Ruiter, G.; Felip, E.; Castanon Alvarez, E.; Ramalingam, S.S.; Rolfo, C.D.; Spigel, D.R.; Andrew, D.; et al. 1313MO Safety and preliminary efficacy of AZD7789, a bispecific antibody targeting PD-1 and TIM-3, in patients (Pts) with stage IIIB–IV non-small-cell lung cancer (NSCLC) with previous anti-PD-(L)1 therapy. Ann. Oncol. 2023, 34, S755. [Google Scholar] [CrossRef]
  196. Hellmann, M.D.; Bivi, N.; Calderon, B.; Shimizu, T.; Delafontaine, B.; Liu, Z.T.; Szpurka, A.M.; Copeland, V.; Hodi, F.S.; Rottey, S.; et al. Safety and immunogenicity of LY3415244, a bispecific antibody against TIM-3 and PD-L1, in patients with advanced solid tumors. Clin. Cancer Res. 2021, 27, 2773–2781. [Google Scholar] [CrossRef] [PubMed]
  197. Creelan, B.C.; Antonia, S.J. The NKG2A immune checkpoint—A new direction in cancer immunotherapy. Nat. Rev. Clin. Oncol. 2019, 16, 277–278. [Google Scholar] [CrossRef]
  198. Cazzetta, V.; Depierreux, D.; Colucci, F.; Mikulak, J.; Mavilio, D. NKG2A immune checkpoint in Vδ2 T Cells: Emerging application in cancer immunotherapy. Cancers 2023, 15, 1264. [Google Scholar] [CrossRef]
  199. Raulet, D.H.; Vance, R.E.; McMahon, C.W. Regulation of the natural killer cell receptor repertoire. Annu. Rev. Immunol. 2001, 19, 291–330. [Google Scholar] [CrossRef]
  200. Kabat, J.; Borrego, F.; Brooks, A.; Coligan, J.E. Role that each NKG2A immunoreceptor tyrosine-based inhibitory motif plays in mediating the human CD94/NKG2A inhibitory signal. J. Immunol. 2002, 169, 1948–1958. [Google Scholar] [CrossRef]
  201. Lanier, L.L. NK CELL RECOGNITION. Annu. Rev. Immunol. 2005, 23, 225–274. [Google Scholar] [CrossRef]
  202. Borst, J.; Ahrends, T.; Bąbała, N.; Melief, C.J.M.; Kastenmüller, W. CD4+ T cell help in cancer immunology and immunotherapy. Nat Rev Immunol 2018, 18, 635–647. [Google Scholar] [CrossRef]
  203. Gemelli, M.; Noonan, D.M.; Carlini, V.; Pelosi, G.; Barberis, M.; Ricotta, R.; Albini, A. Overcoming resistance to checkpoint inhibitors: Natural killer cells in non-small cell lung cancer. Front. Oncol 2022, 12, 886440. [Google Scholar] [CrossRef] [PubMed]
  204. Kyrysyuk, O.; Wucherpfennig, K.W. Designing cancer immunotherapies that engage T cells and NK cells. Annu. Rev. Immunol. 2023, 41, 17–38. [Google Scholar] [CrossRef]
  205. Viant, C.; Fenis, A.; Chicanne, G.; Payrastre, B.; Ugolini, S.; Vivier, E. SHP-1-mediated inhibitory signals promote responsiveness and anti-tumour functions of natural killer cells. Nat. Commun. 2014, 5, 5108. [Google Scholar] [CrossRef] [PubMed]
  206. Niogret, C.; Miah, S.M.S.; Rota, G.; Fonta, N.P.; Wang, H.; Held, W.; Birchmeier, W.; Sexl, V.; Yang, W.; Vivier, E.; et al. Shp-2 is critical for ERK and metabolic engagement downstream of IL-15 receptor in NK cells. Nat. Commun. 2019, 10, 1444. [Google Scholar] [CrossRef]
  207. Yazdi, M.T.; Van Riet, S.; Van Schadewijk, A.; Fiocco, M.; Van Hall, T.; Taube, C.; Hiemstra, P.S.; Van Der Burg, S.H. The positive prognostic effect of stromal CD8+ tumor-infiltrating T cells is restrained by the expression of HLA-E in non-small cell lung carcinoma. Oncotarget 2016, 7, 3477–3488. [Google Scholar] [CrossRef] [PubMed]
  208. Tang, K.H.; Li, S.; Khodadadi-Jamayran, A.; Jen, J.; Han, H.; Guidry, K.; Chen, T.; Hao, Y.; Fedele, C.; Zebala, J.A.; et al. Combined inhibition of SHP2 and CXCR1/2 promotes antitumor T-cell response in NSCLC. Cancer Discov. 2022, 12, 47–61. [Google Scholar] [CrossRef]
  209. Borst, L.; Sluijter, M.; Sturm, G.; Charoentong, P.; Santegoets, S.J.; Van Gulijk, M.; Van Elsas, M.J.; Groeneveldt, C.; Van Montfoort, N.; Finotello, F.; et al. NKG2A is a late immune checkpoint on CD8 T cells and marks repeated stimulation and cell division. Intl J. Cancer 2022, 150, 688–704. [Google Scholar] [CrossRef]
  210. Braud, V.M.; Allan, D.S.J.; O’Callaghan, C.A.; Söderström, K.; D’Andrea, A.; Ogg, G.S.; Lazetic, S.; Young, N.T.; Bell, J.I.; Phillips, J.H.; et al. HLA-E binds to natural killer cell receptors CD94/NKG2A, B and C. Nature 1998, 391, 795–799. [Google Scholar] [CrossRef]
  211. Lanier, L.L.; Corliss, B.; Wu, J.; Phillips, J.H. Association of DAP12 with activating CD94/NKG2C NK cell receptors. Immunity 1998, 8, 693–701. [Google Scholar] [CrossRef]
  212. Vales-Gomez, M. Kinetics and Peptide Dependency of the Binding of the Inhibitory NK Receptor CD94/NKG2-A and the Activating Receptor CD94/NKG2-C to HLA-E. EMBO J. 1999, 18, 4250–4260. [Google Scholar] [CrossRef]
  213. Kaiser, B.K.; Barahmand-pour, F.; Paulsene, W.; Medley, S.; Geraghty, D.E.; Strong, R.K. Interactions between NKG2x immunoreceptors and HLA-E ligands display overlapping affinities and thermodynamics. J. Immunol. 2005, 174, 2878–2884. [Google Scholar] [CrossRef] [PubMed]
  214. Lauterbach, N.; Wieten, L.; Popeijus, H.E.; Voorter, C.E.M.; Tilanus, M.G.J. HLA-E regulates NKG2C+ natural killer cell function through presentation of a restricted peptide repertoire. Hum. Immunol. 2015, 76, 578–586. [Google Scholar] [CrossRef]
  215. Siemaszko, J.; Marzec-Przyszlak, A.; Bogunia-Kubik, K. Activating NKG2C receptor: Functional characteristics and current strategies in clinical applications. Arch. Immunol. Ther. Exp. 2023, 71, 9. [Google Scholar] [CrossRef] [PubMed]
  216. Battin, C.; Kaufmann, G.; Leitner, J.; Tobias, J.; Wiedermann, U.; Rölle, A.; Meyer, M.; Momburg, F.; Steinberger, P. NKG2A -checkpoint Inhibition and its blockade critically depends on peptides presented by its ligand HLA-E. Immunology 2022, 166, 507–521. [Google Scholar] [CrossRef]
  217. Nuttin, L.; Latour, C.; Revy, S.; Aucagne, R.; Lalle, G.; Ghiringhelli, F.; Limagne, E. Abstract 3864: Role of NK/TAM cross-talk on T-CD8+ recruitment and efficacy of chemoimmunotherapy and MEKi combination in “cold” tumors. Cancer Res. 2024, 84, 3864. [Google Scholar] [CrossRef]
  218. André, P.; Denis, C.; Soulas, C.; Bourbon-Caillet, C.; Lopez, J.; Arnoux, T.; Bléry, M.; Bonnafous, C.; Gauthier, L.; Morel, A.; et al. Anti-NKG2A mAb is a checkpoint inhibitor that promotes anti-tumor immunity by unleashing both T and NK cells. Cell 2018, 175, 1731–1743.e13. [Google Scholar] [CrossRef] [PubMed]
  219. Cascone, T.; Kar, G.; Spicer, J.D.; García-Campelo, R.; Weder, W.; Daniel, D.B.; Spigel, D.R.; Hussein, M.; Mazieres, J.; Oliveira, J.; et al. Neoadjuvant durvalumab alone or combined with novel immuno-oncology agents in resectable lung cancer: The phase II NeoCOAST platform trial. Cancer Discov. 2023, 13, 2394–2411. [Google Scholar] [CrossRef]
  220. Herbst, R.S.; Majem, M.; Barlesi, F.; Carcereny, E.; Chu, Q.; Monnet, I.; Sanchez-Hernandez, A.; Dakhil, S.; Camidge, D.R.; Winzer, L.; et al. COAST: An open-label, phase II, multidrug platform study of durvalumab alone or in combination with oleclumab or monalizumab in patients with unresectable, stage III non–small-cell lung cancer. J. Clin. Oncol. 2022, 40, 3383–3393. [Google Scholar] [CrossRef]
  221. Aggarwal, C.; Martinez-Marti, A.; Majem, M.; Barlesi, F.; Carcereny, E.; Chu, Q.S.; Monnet, I.; Sanchez, A.; Dakhil, S.R.; Camidge, D.R.; et al. Updated results from COAST, a phase 2 study of Durvalumab (D) ± Oleclumab (O) or Monalizumab (M) in patients (Pts) with stage III unresectable non-small cell lung cancer (uNSCLC). J. Clin. Oncol. 2024, 42, 8046. [Google Scholar] [CrossRef]
  222. Barlesi, F.; Cho, B.C.; Goldberg, S.B.; Yoh, K.; Zimmer Gelatti, A.C.; Mann, H.; Gopinathan, A.; Bielecka, Z.F.; Newton, M.; Aggarwal, C. PACIFIC-9: Phase III trial of Durvalumab + Oleclumab or Monalizumab in unresectable stage III non-small-cell lung cancer. Future Oncol. 2024, 20, 2137–2147. [Google Scholar] [CrossRef]
  223. Patel, S.P.; Alonso-Gordoa, T.; Banerjee, S.; Wang, D.; Naidoo, J.; Standifer, N.E.; Palmer, D.C.; Cheng, L.Y.; Kourtesis, P.; Ascierto, M.L.; et al. Phase 1/2 study of monalizumab plus durvalumab in patients with advanced solid tumors. J Immunother Cancer 2024, 12, e007340. [Google Scholar] [CrossRef] [PubMed]
  224. Tomasini, P.; Cropet, C.; Jeanson, A.; Pérol, M.; Mazieres, J.; Fontaine, C.; Rabeau, A.; Meurer, M.; Greillier, L.; Le Ray, M.; et al. LBA8 Precision immuno-oncology for advanced non-small cell lung cancer (NSCLC) patients with PD-(L)1 inhibitors resistance (PIONeeR): A phase Ib/IIa clinical trial targeting identified resistance pathways. Ann. Oncol. 2024, 35, S1242. [Google Scholar] [CrossRef]
  225. Melander, M.; Laugel, B. Abstract LB220: S095029: A novel clinical-stage Fc-silenced NKG2A-blocking antibody with best-in-class potential. Cancer Res. 2023, 83, LB220. [Google Scholar] [CrossRef]
  226. Spreafico, A.; Naing, A.; Barve, M.; Patnaik, A.; Ianopulos, X.; Drean, P.; Hemon, A.; Kantari, C.C.; He, P.; Askoxylakis, V.; et al. 1495 A Phase 1 Multicenter Study of the Safety and Efficacy of the NKG2A Targeting Antibody S095029 as Single Agent and in Combination with Anti-PD1 in Patients with Advanced Malignancies. J. ImmunoTherapy Cancer 2024, 12. [Google Scholar] [CrossRef]
  227. Vijayan, D.; Young, A.; Teng, M.W.L.; Smyth, M.J. Targeting immunosuppressive adenosine in cancer. Nat. Rev. Cancer 2017, 17, 709–724. [Google Scholar] [CrossRef]
  228. Allard, B.; Allard, D.; Buisseret, L.; Stagg, J. The adenosine pathway in immuno-oncology. Nat. Rev. Clin. Oncol. 2020, 17, 611–629. [Google Scholar] [CrossRef]
  229. Kaplinsky, N.; Williams, K.; Watkins, D.; Adams, M.; Stanbery, L.; Nemunaitis, J. Regulatory role of CD39 and CD73 in tumor immunity. Future Oncol. 2024, 20, 1367–1380. [Google Scholar] [CrossRef]
  230. Resta, R.; Yamashita, Y.; Thompson, L.F. Ecto-enzyme and Signaling Functions of Lymphocyte CD 7 3. Immunological Reviews 1998, 161, 95–109. [Google Scholar] [CrossRef]
  231. Liu, D.; Zhao, J.; Li, L.; Wang, J.; Wang, C.; Wu, Y.; Huang, Y.; Xing, D.; Chen, W. CD73: Agent development potential and its application in diabetes and atherosclerosis. Front. Immunol. 2024, 15, 1515875. [Google Scholar] [CrossRef]
  232. Allard, B.; Longhi, M.S.; Robson, S.C.; Stagg, J. The ectonucleotidases CD 39 and CD 73: Novel checkpoint inhibitor targets. Immunol. Rev. 2017, 276, 121–144. [Google Scholar] [CrossRef]
  233. Imbimbo, M.; Hollebecque, A.; Italiano, A.; McKean, M.; Macarulla, T.; Castanon Alvarez, E.; Carneiro, B.A.; Mager, R.; Barnhart, V.; Murtomaki, E.; et al. 188P IPH5201 as monotherapy or in combination with durvalumab (D) in advanced solid tumours. Immuno. Oncol. Technol. 2022, 16, 100300. [Google Scholar] [CrossRef]
  234. Barlesi, F.; Mercier, O.; Łowczak, A.; Mandziuk, S.; Kuzdzal, J.; Koutras, A.; Dasgupta, A.; Leyco, A.J.; Andre, P.; Mager, R.; et al. 1290TiP a phase II multicenter, open label, non-randomized study of neoadjuvant and adjuvant treatment with IPH5201 and durvalumab in patients with resectable, early-stage (II to IIIA) non-small cell lung cancer (MATISSE). Ann. Oncol. 2023, 34, S744. [Google Scholar] [CrossRef]
  235. Wainberg, Z.A.; Cha, Y.; George, B.; Grewal, J.S.; Merchan, J.R.; Mckean, M.; Soares, H.P.; Cruz, T.D.; Moesta, A.K.; Jahn, T.M.; et al. 659P combination treatment with TTX-030, a first-in-class anti-CD39 antibody, in patients with advanced pancreatic cancer. Ann. Oncol. 2024, 35, S519–S520. [Google Scholar] [CrossRef]
  236. Thompson, L.F.; Ruedi, J.M.; Glass, A.; Low, M.G.; Lucas, A.H. Antibodies to 5’-nucleotidase (CD73), a glycosyl-phosphatidylinositol-anchored protein, cause human peripheral blood T cells to proliferate. J. Immunol. 1989, 143, 1815–1821. [Google Scholar] [CrossRef]
  237. Wang, L.; Fan, J.; Thompson, L.F.; Zhang, Y.; Shin, T.; Curiel, T.J.; Zhang, B. CD73 has distinct roles in nonhematopoietic and hematopoietic cells to promote tumor growth in mice. J. Clin. Invest. 2011, 121, 2371–2382. [Google Scholar] [CrossRef]
  238. Stagg, J.; Beavis, P.A.; Divisekera, U.; Liu, M.C.P.; Möller, A.; Darcy, P.K.; Smyth, M.J. CD73-deficient mice are resistant to carcinogenesis. Cancer Res. 2012, 72, 2190–2196. [Google Scholar] [CrossRef]
  239. Turcotte, M.; Spring, K.; Pommey, S.; Chouinard, G.; Cousineau, I.; George, J.; Chen, G.M.; Gendoo, D.M.A.; Haibe-Kains, B.; Karn, T.; et al. CD73 is associated with poor prognosis in high-grade serous ovarian cancer. Cancer Res. 2015, 75, 4494–4503. [Google Scholar] [CrossRef]
  240. Allard, B.; Turcotte, M.; Spring, K.; Pommey, S.; Royal, I.; Stagg, J. Anti-CD73 therapy impairs tumor angiogenesis. Int. J. Cancer 2014, 134, 1466–1473. [Google Scholar] [CrossRef]
  241. Bendell, J.; LoRusso, P.; Overman, M.; Noonan, A.M.; Kim, D.-W.; Strickler, J.H.; Kim, S.W.; Clarke, S.; George, T.J.; Grimison, P.S.; et al. First-in-human study of oleclumab, a potent, selective anti-CD73 monoclonal antibody, alone or in combination with durvalumab in patients with advanced solid tumors. Cancer Immunol. Immunother 2023, 72, 2443–2458. [Google Scholar] [CrossRef]
  242. Fredholm, B.B.; IJzerman, A.P.; Jacobson, K.A.; Linden, J.; Müller, C.E. international union of basic and clinical pharmacology. LXXXI. Nomenclature and classification of adenosine receptors—An update. Pharmacol. Rev. 2011, 63, 1–34. [Google Scholar] [CrossRef]
  243. Chiappori, A.A.; Creelan, B.; Tanvetyanon, T.; Gray, J.E.; Haura, E.B.; Thapa, R.; Barlow, M.L.; Chen, Z.; Chen, D.T.; Beg, A.A.; et al. Phase I study of taminadenant (PBF509/NIR178), an adenosine 2A receptor antagonist, with or without spartalizumab (PDR001), in patients with advanced non–small cell lung cancer. Clin. Cancer Res. 2022, 28, 2313–2320. [Google Scholar] [CrossRef] [PubMed]
Figure 1. CTLA-4 mechanism of action. APC: antigen-presenting cell.
Figure 1. CTLA-4 mechanism of action. APC: antigen-presenting cell.
Cancers 17 00906 g001
Figure 2. TIGIT mechanism of action. APC: antigen-presenting cell.
Figure 2. TIGIT mechanism of action. APC: antigen-presenting cell.
Cancers 17 00906 g002
Figure 3. LAG-3 mechanism of action. APC: antigen-presenting cell; FGL1: fibrogen-like protein 1; Gal-3: galectin-3; sLAG-3: soluble LAG-3.
Figure 3. LAG-3 mechanism of action. APC: antigen-presenting cell; FGL1: fibrogen-like protein 1; Gal-3: galectin-3; sLAG-3: soluble LAG-3.
Cancers 17 00906 g003
Figure 4. TIM-3 mechanism of action. APC: antigen-presenting cell; Gal-9: galectin-9; PtdSer: phosphatidylserine.
Figure 4. TIM-3 mechanism of action. APC: antigen-presenting cell; Gal-9: galectin-9; PtdSer: phosphatidylserine.
Cancers 17 00906 g004
Figure 5. NKG2A mechanism of action. APC: antigen-presenting cell.
Figure 5. NKG2A mechanism of action. APC: antigen-presenting cell.
Cancers 17 00906 g005
Figure 6. CD39/CD73/adenosine pathway. Adenosine R: adenosine receptor; AMP: adenosine monophosphate; ADP: adenosine diphosphate; ATP: adenosine triphosphate; MDSC: myeloid-derived suppressor cell.
Figure 6. CD39/CD73/adenosine pathway. Adenosine R: adenosine receptor; AMP: adenosine monophosphate; ADP: adenosine diphosphate; ATP: adenosine triphosphate; MDSC: myeloid-derived suppressor cell.
Cancers 17 00906 g006
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Roussot, N.; Kaderbhai, C.; Ghiringhelli, F. Targeting Immune Checkpoint Inhibitors for Non-Small-Cell Lung Cancer: Beyond PD-1/PD-L1 Monoclonal Antibodies. Cancers 2025, 17, 906. https://doi.org/10.3390/cancers17050906

AMA Style

Roussot N, Kaderbhai C, Ghiringhelli F. Targeting Immune Checkpoint Inhibitors for Non-Small-Cell Lung Cancer: Beyond PD-1/PD-L1 Monoclonal Antibodies. Cancers. 2025; 17(5):906. https://doi.org/10.3390/cancers17050906

Chicago/Turabian Style

Roussot, Nicolas, Courèche Kaderbhai, and François Ghiringhelli. 2025. "Targeting Immune Checkpoint Inhibitors for Non-Small-Cell Lung Cancer: Beyond PD-1/PD-L1 Monoclonal Antibodies" Cancers 17, no. 5: 906. https://doi.org/10.3390/cancers17050906

APA Style

Roussot, N., Kaderbhai, C., & Ghiringhelli, F. (2025). Targeting Immune Checkpoint Inhibitors for Non-Small-Cell Lung Cancer: Beyond PD-1/PD-L1 Monoclonal Antibodies. Cancers, 17(5), 906. https://doi.org/10.3390/cancers17050906

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop