Next Article in Journal
The Role of Cytokines in the Development and Functioning of the Hypothalamic–Pituitary–Gonadal Axis in Mammals in Normal and Pathological Conditions
Previous Article in Journal
The Paraoxonase (PON) Gene Family in Health, Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) and Other Diseases
Previous Article in Special Issue
Hematological Biomarkers for Early Detection of Lung Cancer: Evaluating the Diagnostic Potential of Circulating Interleukin Levels
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Directions of Immunotherapy for Non-Small-Cell Lung Cancer Treatment: Past, Present and Possible Future

Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95123 Catania, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(22), 11055; https://doi.org/10.3390/ijms262211055 (registering DOI)
Submission received: 16 October 2025 / Revised: 11 November 2025 / Accepted: 13 November 2025 / Published: 15 November 2025

Abstract

Lung cancer (LC) is one of the most common malignancies and the leading cause of death worldwide. LC is classified into two main histological subtypes: non-small-cell lung cancer (NSCLC), representing the 85% of all LC types, and small-cell lung cancer (SCLC), representing 15% of all lung neoplasm. The recent discovery and clinical approval of new therapeutic approaches has resulted in significant advancements in the management of NSCLC patients. This review aims to summarize the current and ongoing clinical trials that have led to the approval of immune checkpoint inhibitors (ICIs) and the emerging immunotherapy approaches for advanced NSCLC patients. Additionally, the current benefits and drawbacks of these therapeutic strategies will be explored. The treatment for NSCLC is evolving toward a more comprehensive approach that considers both the tumor immune history and genomic features. In this respect, we hope that the ongoing research will make it possible to treat each NSCLC patient individually in the near future.

1. Introduction

Lung cancer (LC) is one of the most common malignancies and the leading cause of death worldwide for cancer, in both sexes [1]. Despite the development of new advanced therapies, LC is still a malignancy with an unfavorable prognosis, typically due to the non-specific early symptoms and the lack of a reliable screening test [2,3,4,5].
LC is classified into lung microcitoma or small-cell lung cancer (SCLC) and non-small-cell lung cancer (NSLC), which includes squamous or epidermoid carcinoma (SqCC), adenocarcinoma (ADC), and large cell carcinoma (LCC) [6]. NSCLCs represent around 85% of all LC types, while SCLCs is less common (approximately 15% of lung neoplasms) but characterized by faster growth and a higher propensity for metastasis [7,8,9]. Additionally, rare forms of LC include: adeno-squamous carcinoma, salivary gland-type lung carcinoma, large cell neuroendocrine carcinoma and lung carcinoids [6,10,11].
The gold standard treatment for LC varies based on the type, stage, and molecular profile of the tumor, as well as the patient overall health and preferences [12]. For early-stage NSCLC, surgical resection remains the primary treatment, often followed by adjuvant chemotherapy or radiotherapy to reduce the risk of recurrence [13]. LC outcomes are strongly influenced by the stage at diagnosis. Patients diagnosed at an early stage generally have a favorable prognosis, with 5-year survival rates of approximately 70–90% for Stage I and 50–70% for Stage II [14]. However, survival decreases markedly in more advanced stages, reaching 30–35% for Stage III and only about 9–10% for Stage IV. Unfortunately, LC is most often diagnosed at an advanced stage, when curative options are limited and systemic drug therapy, guided by the immunological and molecular profile of the tumor, represents the standard of care [15,16,17].
Over the last decades, numerous evidence have progressively led to a better understanding of the interactions between cancer cells and the immune system, emphasizing the key role of the immune system in suppressing the development and progression of cancer. The immune response is able to control the growth of tumor cells. In fact, the presence of lymphocytes infiltrating tumor (TILs) is often associated with longer patient survival [18,19]. However, some tumors, known as “cold tumors”, are particularly resistant to the immune response. In particular, these are characterized by the lack of recognizable mutations, an immunosuppressive tumor microenvironment (TME), and the expression of proteins that inhibit the action of T lymphocytes [20].
The advent of immunotherapy has revolutionized the treatment landscape for LC, particularly NSCLC [21,22]. Immune checkpoint inhibitors (ICIs), such as those targeting the PD-1 (programmed death-1) receptor and its ligand PD-L1, have already demonstrated to provide significant improvements in survival for patients with advanced or metastatic NSCLC [12,23]. Drugs like ipilimumab, tremelimumab, pembrolizumab, nivolumab, durvalumab, cemiplimab, and atezolizumab have received both the Food and Drug Administration (FDA) approval for use in various stages of the disease, both as monotherapy and in combination with chemotherapy (Figure 1) [24,25,26,27,28]. These agents work by enhancing the antitumor immune response, releasing the inhibition of T cells exerted by the tumor [29,30]. However, responses are not universal, and primary or acquired resistance and the absence of predictive markers of response remain a challenge [31,32]. Additionally, the optimization of therapeutic combinations and the management of side effects are areas of ongoing research studies. Recent studies are focusing on identifying predictive biomarkers, such as PD-L1 expression, tumor mutational burden (TMB) and Tumor-infiltrating lymphocytes (TILs), and on optimizing therapeutic combinations to further improve the outcomes and to reduce adverse drug reactions [33,34,35]. However, despite their central role in guiding immunotherapy, these biomarkers showed significant limitations. In particular, PD-L1 expression is highly heterogeneous both spatially and temporally, varying between primary and metastatic sites and even within the same tumor region [36]. Its assessment is further complicated by inter-assay variability among immunohistochemical tests (e.g., 22C3, 28-8, SP263, SP142) and the lack of standardized cutoff values, which were historically defined in trial-specific contexts rather than through biologically consistent thresholds [37]. Similarly, TMB, though associated with improved response to immune checkpoint inhibitors, lacks a universally accepted assay or cutoff, and its predictive value may differ depending on sequencing platform, bioinformatic pipeline, and tumor subtype [38,39]. Moreover, TMB does not fully capture the immunogenic landscape, since not all mutations generate neoantigens capable of eliciting an immune response [40]. The use of TILs as a biomarker also faces methodological challenges due to subjective interpretation, intratumoral variability, and absence of validated quantification standards in lung cancer [41]. Collectively, these factors limit the reliability and reproducibility of current biomarkers and underscore the need for integrated, multi-parametric approaches combining molecular, histopathological, and immunological data to better predict immunotherapy outcomes in NSCLC.
This review explores the rationale and current evidence supporting approved immunotherapy strategies for advanced and metastatic NSCLC, integrating insights from preclinical research and ongoing clinical trials. Particular attention is given to emerging combination strategies aimed at overcoming resistance and improving clinical outcomes. Furthermore, novel therapeutic approaches, current challenges, and future perspectives in the evolving landscape of NSCLC immunotherapy will be discussed.

2. Immune Checkpoint Inhibitors

Although the immune system and cancer cells coexist, the persistence of these can lead to a depletion of effective anti-tumor T-lymphocytes (“T-cell exhaustion”). In particular, T cells undergo a functional paralysis (“anergy”) leading to the expression of inhibitory immune-checkpoints on their surface, which are engaged by the tumor cells to suppress the immune response [42].
The progress in the understanding of the immunobiology, combined with the recent discoveries on physiological mechanisms that regulate the activity of the immune system and its interactions with the tumor, have allowed development of a new class of therapeutic agents represented by immunomodulatory mAbs capable of blocking ICIs.
Among the various immune checkpoints leveraged by tumors to evade the host immune system, the PD-1/PD-L1 and CTLA-4 pathways are the most well-studied and advanced in clinical applications. Blocking these pathways restores the priming and anti-tumor activity of cytotoxic T cells, which are otherwise inhibited.

2.1. PD-1/PD-L1 Axis

The first major breakthrough in the use of ICIs for NSCLC was the FDA approval of nivolumab (a human IgG4 monoclonal antibody (mAb) targeting PD-1) as a second-line therapy for advanced and metastatic NSCLC, in 2015. The approval was based on the results of CheckMate 017 (for SqCC patients) and CheckMate 057 (for non-SqCC patients) phase III trials (Table 1) [26,43]. These trials demonstrated that nivolumab provided superior objective response rates (ORRs) in patients with SqCC and non-SqCC, who had progressed following platinum-based chemotherapy (20% and 19%, respectively) compared to docetaxel group (12.4%). Additionally, patients treated with nivolumab showed a better median OS (9.2 and 12.2 months, respectively) than docetaxel group (6.0 and 9.4 months, respectively), with a 27% to 41% reduction in mortality risk (for non-SqCC and SqCC patients, respectively) [26,43]. Although the studies represented a milestone in shifting NSCLC treatment toward immunotherapy, several limitations should be acknowledged. Biomarker analysis showed that PD-L1 expression was not predictive of clinical benefit in this population, underscoring the complexity of immune response mechanisms and the need for more accurate predictive biomarkers. Additionally, the study design involved a comparison with docetaxel, a drug that is now largely outdated, thereby reducing the long-term relevance of the trial as a reference.
Along the same lines, other ICIs such as pembrolizumab (a humanized IgG4 anti-PD-1 mAb) and atezolizumab (a humanized IgG1 anti-PD-L1 mAb), marked a second breakthrough in the treatment of NSCLC due to their demonstrated efficacy over docetaxel in second-line settings. The success of ICIs in second-line treatment paved the way for their use as first-line therapy for advanced NSCLC.
At the end of 2015, the FDA approved pembrolizumab for the treatment of patients with advanced or metastatic NSCLC expressing PD-L1, following disease progression after platinum-based chemotherapy [44]. Subsequently, in 2016, it was approved as a first-line therapy for patients with advanced NSCLC with PD-L1 expression ≥ 50%, based on the results of the randomized phase III study KEYNOTE-024 (Table 1) [45]. This trial demonstrated that patients treated with pembrolizumab showed a significant improvement in OS rate at 6 months (80.2%) and progression-free survival (PFS) (10.3 months) compared to standard chemotherapy group (72.4% and 6 months, respectively). In addition, Pembrolizumab also demonstrated a higher response rate (44.8% vs. 27.8%) and less frequent adverse drug reaction (73.4% vs. 90.0%) [45]. However, the study population was highly selected, excluding patients with PD-L1 < 50% and those with targetable mutations, which limits generalizability to the broader NSCLC population. Moreover, the reliance on PD-L1 as a single biomarker overlooks the complexity of tumor immunogenicity and the influence of other factors such as tumor mutational burden (TMB) and immune microenvironment. Finally, although long-term follow-up confirmed durable benefit, primary resistance and late relapses remain significant challenges.
Similarly, atezolizumab received the first FDA approval for previously treated patients with metastatic NSCLC, in 2016. The approval was based on the results of the OAK trial (Table 1) [46]. This trial demonstrated that atezolizumab provided a superior OS (15.7 months) compared with chemotherapy alone (10.3 months). In addition, both SqCC and non-SqCC patients with higher PD-L1 expression treated with atezolizumab experienced a longer median OS (20.5 months) [46].
Two years later, a new fully human IgG1 mAb targeting PD-L1 (durvalumab) received the FDA approval to treat patients with unresectable advanced NSCLC, who were previously treated with chemoradiation [47]. Indeed, the results of the PACIFIC study demonstrated that durvalumab increase the median OS (47.5 months versus 29.1 months of placebo) with a 4-year OS rate of 49.6% (for durvalumab arm) compared with 36.3% (for placebo group), and 4-year progression-free survival (PFS) rate of 35.3% compared with 19.5%, respectively (Table 1) [48]. The approval has been extended over time, including combinations with tremelimumab (a human IgG2 mAb targeted CTLA-4) plus chemotherapy for patients with metastatic NSCLC, as demonstrated in POSEIDON, phase III trial [49]. These studies supports durvalumab as a viable first-line option for metastatic NSCLC, particularly in PD-L1–low or negative patients, but its incremental benefit and optimal patient selection require further clarification in future comparative studies. In particular, the magnitude of benefit was modest compared with other contemporary trials, and no clear biomarker-driven subgroup showed a dramatically superior outcome. The addition of tremelimumab increased immune-related adverse events (irAE), raising concerns about the balance between efficacy and toxicity. Additionally, the exclusion of patients with disease progression or poor performance status limits the applicability of results to real-world populations. Moreover, in the evolving landscape of immuno-chemotherapy combinations, the clinical positioning of the durvalumab–tremelimumab regimen remains uncertain, as more established combinations (e.g., nivolumab + ipilimumab) have shown comparable or superior long-term results.
Finally, a human IgG4 mAb targeted PD-1 (cemiplimab) has been approved for the treatment of advanced or metastatic NSCLC with PD-L1 expression ≥ 50%, based on the results of the phase III, EMPOWER-Lung 1 trial, in 2021 (Table 1) [50]. The study compared cemiplimab monotherapy to standard first-line chemotherapy in patients without driver genetic alterations, such as EGFR or ALK. The results demonstrated a significant improvement in OS, with a hazard ratio (HR) of 0.57 (95% CI: 0.42–0.77), indicating a 43% reduction in the risk of death compared to chemotherapy. Additionally, PFS was superior in the cemiplimab group, with an HR of 0.54 (95% CI: 0.43–0.68) [50]. The ORR was 37% in cemiplimab group compared to 21% in patient treated with chemotherapy. One year later, the results from the EMPOWER-Lung 3 phase III study provided further support for the routine administration of cemiplimab plus platinum-based chemotherapy, as a first-line option for NSCLC with high PD-L1 expression (Table 1) [51]. Although the EMPOWER-Lung 3 trial demonstrated significant improvements in OS and PFS with cemiplimab plus chemotherapy, several limitations should be considered. The study included patients with any level of PD-L1 expression, but it was not powered to provide definitive conclusions for subgroups such as PD-L1–negative patients, leaving uncertainty about the magnitude of benefit in these populations [52]. Additionally, early trial termination for efficacy, while ethically justified, may introduce bias and affect the maturity of the long-term data. Although quality-of-life outcomes favored the combination, the magnitude of improvement was modest, and the trial was not primarily designed to assess these endpoints. Finally, longer follow-up is needed to fully characterize the durability of responses and the long-term safety profile, particularly regarding irAE that may emerge with extended treatment.
However, despite the clinical success of ICIs reported above, most NSCLC patients, treated with a single anti PD-1/PD-L1 agent still fail to initially respond to treatment (primary resistance) or experience disease progression after an initial response (acquired resistance). The potential mechanisms underlying these two types of resistances include the lack of immune cell infiltration in the TME, aberrant PD-L1 expression, and genetic or epigenetic alterations that affect tumor antigen presentation [53,54,55]. Additionally, some tumor cells may exploit alternative immune inhibition pathways or create an immunosuppressive microenvironment [56]. To address these challenges, research is focusing on innovative strategies, such as combining ICIs with targeted therapies, anti-angiogenic agents, chemotherapy, or novel ICIs or immunotherapies strategies, to overcome resistance mechanisms and enhance treatment efficacy [53,56].
Table 1. Overview of Clinical Trial Designs.
Table 1. Overview of Clinical Trial Designs.
StudyEnrollmentEndpointsPhaseRef.
AMBER219Safety and tolerability
Maximum tolerated dose (MTD)
Recommended phase II dose (RP2D)
Phase 1[57]
CA184-156954OS, PFS, ORR, irPFS and irAEsPhase 3[58]
CHECKMATE-9LA719OS, PFS, ORR and DORPhase 3[59,60]
CheckMate-017272OS, PFS, ORR and DORPhase 3[26]
CHECKMATE-2271739OS, PFS, ORR and DORPhase 3[61]
CITYSCAPE135OS, PFS, ORR, DOR and AEPhase 2/3[62]
EMPOWER-Lung 1712OS and PFSPhase 3[51]
EMPOWER-Lung 3466OS, PFS and ORRPhase 3[52]
HARMONi-2398PFSPhase 3[63]
Impower132578OS, PFS, ORR and DORPhase 3[64]
KEYNOTE-024305OS and PFSPhase 3[45]
KEYNOTE-189616OS and PFSPhase 3[65]
KEYNOTE-407559OS and PFSPhase 3[66]
NEOpredict-Lung60Surgery within 43 days, OS and DFSPhase 2[67]
OAK1225OSPhase 3[46]
RELATIVITY-047714OS, PFS and ORRPhase 3[68]
SKYSCRAPER-01534OS and PFSPhase 3[69]

2.2. CTLA-4 Pathway

Cytotoxic T-Lymphocyte Antigen-4 (CTLA-4) is a key immune checkpoint that regulates the amplitude and duration of T-cell activation. It competes with the co-stimulatory receptor CD28 for binding to B7 ligands (CD80/CD86) on antigen-presenting cells (APCs), thereby limiting T-cell proliferation and preventing excessive immune activation [70]. Ipilimumab, a monoclonal antibody targeting CTLA-4, was the first immune checkpoint inhibitor (ICI) to receive FDA approval, demonstrating significant clinical benefit in patients with advanced melanoma compared to the gp100 peptide vaccine [70]. Another anti-CTLA-4 antibody, tremelimumab, remains under clinical evaluation for several solid tumors, including LC [71]. By blocking the CTLA-4/B7 interaction, these antibodies enhance T-cell activation and cytokine production, thereby promoting cytotoxic activity within the TME [72].
Following the success of ipilimumab in melanoma, several trials explored its potential role in NSCLC and SCLC. A Phase II randomized, double-blind study evaluated ipilimumab in combination with chemotherapy as a first-line treatment for both NSCLC and SCLC [73]. The study demonstrated modest improvements in immune-related progression-free survival (irPFS) and OS compared to chemotherapy alone (median OS 12.2 vs. 8.3 months). However, these improvements did not consistently translate into statistically significant clinical benefit, and toxicity remained a concern. In particular, irAEs of grade 3–4 occurred in 15–20% of patients receiving ipilimumab.
In contrast, the Phase III CA184-156 trial yielded disappointing results (Table 1). Among 476 patients with advanced SCLC treated with ipilimumab plus chemotherapy, no significant improvement was observed in either OS (11.0 vs. 10.9 months) or PFS (4.6 vs. 4.4 months) compared with placebo [58]. Moreover, 27% of patients experienced treatment-related immune toxicities, underscoring the narrow therapeutic window of CTLA-4 blockade in this setting.
Collectively, these studies highlight a translational gap between the early promise of CTLA-4 inhibition observed in melanoma and its limited success in lung cancer. Although ipilimumab and tremelimumab remain under investigation in combination with PD-1/PD-L1 inhibitors and chemotherapy, the risk–benefit ratio appears less favourable in unselected NSCLC and SCLC populations. The lack of significant survival benefit in pivotal Phase III trials suggests that CTLA-4 inhibition alone may not be sufficient to overcome the profound immunosuppressive milieu of lung tumors.
Future research should focus on biomarker-driven patient selection, optimal dosing and sequencing strategies, and combination regimens that balance efficacy with manageable toxicity. A deeper understanding of TME heterogeneity, T-cell exhaustion, and irAE mechanisms will be essential to refine the therapeutic positioning of CTLA-4 blockade within the evolving landscape of lung cancer immunotherapy.

3. Combination Therapy

Monotherapy with ICIs has significantly improved survival outcomes for patients with NSCLC, revolutionizing the treatment of these patients. However, several patients do not benefit significantly from immunotherapy alone [74]. For these patients, it has been hypothesized that chemotherapy, traditionally used to reduce tumor burden by inducing DNA damage and apoptosis in tumor cells, may play a key role in modifying the TME [75]. In this context, chemotherapy acts not only as a cytotoxic agent but also as a catalyst that renders the tumor immunogenic, thereby optimizing the efficacy of ICIs.
The understanding of the synergistic potential between chemotherapy and ICIs provided the theoretical foundation for the design of several studies including the two trials KEYNOTE-189 and KEYNOTE-407, which revolutionized first-line treatment for patients with metastatic NSCLC (Table 1). These studies evaluated the efficacy of combining pembrolizumab with standard chemotherapy regimens, demonstrating significant improvements in OS and PFS compared to chemotherapy alone, regardless of PD-L1 expression levels [65,66].
In particular, the KEYNOTE-189 trial included patients with non-SqCC, using a combination of pembrolizumab with pemetrexed and platinum-based chemotherapy, while KEYNOTE-407 focused on patients with SqCC, combining pembrolizumab with carboplatin and paclitaxel (or nab-paclitaxel). The results of both studies demonstrated that the chemotherapy–pembrolizumab combination not only improved the duration of response (DOR) but also significantly reduced the risk of death by 44%, and doubled PFS, extending it from 4.9 months to 9 months [65,66]. Importantly, the observed benefits were independent of PD-L1 expression levels, demonstrating efficacy across all subgroups, including those with PD-L1 < 1%, PD-L1 between 1–49%, and PD-L1 ≥ 50%. These landmark results led to the FDA approval of the chemotherapy–pembrolizumab combination for first-line treatment, prompting updates to international guidelines and establishing this approach as the new standard of care for patients with metastatic NSCLC, in 2018 [76]. Despite the trial clear clinical impact, several limitations should be acknowledged. The study excluded patients with targetable driver mutations and those with poor performance status, limiting the generalizability of the results to these populations. While PD-L1 expression was assessed, the trial was not powered to determine definitive benefits in PD-L1–low or negative subgroups. Furthermore, the control arm consisted of chemotherapy alone, which, given the current availability of multiple immunotherapy combinations, reduces its contemporary relevance. Finally, although long-term follow-up demonstrated durable survival benefits, extended monitoring is needed to fully characterize late irAE and long-term safety.
On the other hand, the results of the Impower132 clinical study demonstrated that patients treated with atezolizumab in combination with carboplatin/cisplatin and paclitaxel showed significant benefits in PFS with a median PFS of 7.6 months compared to 5.2 months for chemotherapy alone (Table 1) [64]. However, the trial did not demonstrate a statistically significant improvement in OS, with median OS reaching 18.1 months in the combined arm versus 13.6 months in the chemotherapy group. Despite the lack of OS benefit, the combination was well tolerated, with manageable safety profiles consistent with those of the individual agents [64,77]. However, the health-related quality-of-life data were limited, making it difficult to assess the broader patient-centered impact of the treatment. Finally, the study did not explore optimal sequencing strategies for patients who progress after the combination, leaving unanswered questions about subsequent therapy choices.
Several randomized trials have demonstrated that combining anti-CTLA-4 with anti-PD-1 mAbs offers greater clinical benefits compared to chemotherapy or single-agent therapies. In particular, among these two randomized clinical trials (CHECKMATE-227 and CHECKMATE-9LA) showed better efficacy of ipilimumab (a fully human IgG1 mAb targeting CTLA-4) plus nivolumab treatment compared with chemotherapy alone, allowing the FDA approval of the dual checkpoint blockade to treat patients with advanced NSCLC patients, in 2020 (Table 1) [78].
In particular, the CHECKMATE-9LA investigated the efficacy of ipilimumab plus nivolumab and two cycles of chemotherapy versus chemotherapy alone [59]. NSCLC patients treated with the combination therapy showed a median OS rate of 14.1 months compared with 10.7 months of patients treated with chemotherapy alone, with an overall survival rate of 38% versus 25%, respectively [59]. Further analyses showed that after a minimum follow-up of 68.6 months, the combination regimen demonstrated a sustained OS benefit compared with chemotherapy alone (HR = 0.74; 95% CI 0.63–0.87), with 6-year OS rates of 16% vs. 10%, respectively. The survival advantage was consistent across PD-L1 expression levels (<1%: 20% vs. 7%; ≥1%: 15% vs. 10%) and histologic subtypes (SqCC: 14% vs. 5%; non-SqCC: 17% vs. 12%) [60]. The 6-year PFS rates were 9% in the nivolumab plus ipilimumab arm versus 3% with chemotherapy alone (HR = 0.70; 95% CI 0.59–0.82). The DOR was notably longer with the immunotherapy-based regimen, with 19% of patients maintaining a response at 6 years, whereas all responses in the chemotherapy arm had ceased by that time. Exploratory genomic analyses indicated that patients harboring KRAS, STK11, KEAP1, or TP53 mutations also derived clinical benefit, confirming the durable efficacy and manageable safety profile of nivolumab plus ipilimumab combined with limited chemotherapy as a first-line regimen for metastatic NSCLC [60].
On the other hand, the CHECKMATE-227 trial evaluated the safety profile of nivolumab plus ipilimumab as first-line treatment compared to standard chemotherapy. The study was conducted in two parts, focusing on patients with high TMB and those stratified by PD-L1 expression [61]. In the cohort with high TMB, the combination treatment demonstrated a significant improvement in OS compared to chemotherapy, with a median OS of 17.1 months versus 14.9 months, respectively [61]. Moreover, long-term follow-up showed durable survival benefits, with a 5-year OS rate of 24% in the combination arm compared to 14% with chemotherapy [79]. Overall, the trial provided landmark evidence supporting dual checkpoint inhibition in advanced NSCLC and expanded understanding of immunotherapy beyond PD-L1 high patients, but its complex design, heterogeneous outcomes, and safety considerations highlight the need for careful patient selection and further studies to optimize therapeutic strategies.
A recent pooled analysis of the two previously described phase 3 studies, highlighted the long-term benefits of first-line treatment with nivolumab plus ipilimumab, with or without two cycles of chemotherapy, compared to chemotherapy alone in patients with metastatic NSCLC and low PD-L1 expression (<1%). The results confirm that this immunotherapy combination offers a durable and clinically meaningful benefit in a population with still unmet therapeutic needs. Indeed, among the 322 patients treated with the combination immunotherapy, the median OS was 17.4 months, significantly longer than the 11.3 months observed in the chemotherapy arm (HR = 0.64), with a 5-year OS rate of 20% versus 7%. The benefit was consistent across particularly challenging subgroups, such as patients with brain metastases (HR = 0.44) or squamous NSCLC (HR = 0.51). In the overall population, the median PFS was 5.4 months with nivolumab/ipilimumab, compared to 4.9 months with chemotherapy, and the median DOR was 18.0 months versus 4.6 months [80].
Efforts are also underway to combine immune checkpoint inhibitors (ICIs) with targeted therapies, including FDA-approved anti-VEGF monoclonal antibodies (mAbs) such as ranibizumab and bevacizumab. These agents are being explored for their potential to enhance immunomodulation while exerting both anti-angiogenic and immune-modulating effects on tumor cells. This combinatorial approach aims to investigate potential additive or synergistic effects, improving therapeutic efficacy against tumors [81,82,83].
The use of bevacizumab in the treatment of lung neoplasms could represent a valid therapeutic opportunity to overcome tumor resistance to monotherapy [84]. Indeed, anti-VEGF therapies help stabilize newly formed blood vessels within the tumor, reducing abnormal vascular permeability. This stabilization enhances the infiltration of lymphocytes into the TME, potentially improving the efficacy of immune responses and immunotherapy [85].
Although the ICIs represent an effective treatment on a broad spectrum of NSCLC, their utility in the therapeutic algorithm for “oncogenic-addicted” tumors is yet to be demonstrated. However, it has been reported that the overexpression of PD-L1 with TPS (Tumor Proportion Score) > 50% of tumor cells can be observed in tumors with ROS1 rearrangement and, less frequently, in tumors with rearrangements of ALK [86,87,88]. Occasionally, EGFR-mutated tumors may also exhibit marked expression of PD-L1 [89]. In these cases, the overexpression of PD-L1 does not allow the eligibility of the patient for ICI, since the latter may be ineffective and inhibitors of great benefit tyrosine kinases. Overall, it is clear how the assessment of PD-L1 expression should be included as part of a more elaborate diagnostic/therapeutic algorithm.

4. Novel Immune Checkpoint Targets

Although ICIs have revolutionized the treatment of NSCLC, there remains significant potential to enhance their efficacy and broaden their utility. Despite their success in a subset of patients, many individuals remain ineligible, fail to respond, or eventually develop resistance to therapies targeting CTLA-4 and PD-1/PD-L1 pathways [90].
Resistance to CTLA-4 or PD-1/PD-L1 blockades remains a major barrier in the treatment of NSCLC. A significant proportion of patients exhibit primary resistance, meaning they derive minimal or no benefit from the outset, while others develop acquired resistance after an initial response. Mechanistically, primary resistance often stems from tumor–immune escape at multiple levels of the cancer–immunity cycle: deficient antigen presentation (e.g., downregulation of HLA-I or loss of β2-microglobulin), oncogenic pathway activation (e.g., STK11, KEAP1, EGFR/ALK aberrations) and an immunosuppressive microenvironment with low T-cell infiltration or high myeloid-derived suppressor cell (MDSC) load [91].
On the other hand, acquired resistance may result from clonal selection of tumor sub-populations lacking neoantigens, epigenetic or signaling pathway changes (e.g., JAK1/JAK2 mutations, IFN-γ pathway defects), upregulation of alternative immune checkpoints (e.g., TIM-3, LAG-3) and remodelling of the tumor microenvironment to a “cold” phenotype. Translational investigations in NSCLC have, for example, documented acquired loss of HLA class I expression with matched pre- and post-treatment biopsies, and reduced CD8+ T-cell infiltration at progression [92]. These mechanistic insights are now informing therapeutic strategies to overcome resistance: explore and identify alternative immune checkpoints, such as TIGIT, LAG-3, and TIM-3, dual checkpoint blockade (CTLA-4 + PD-1 or PD-1 + LAG-3/TIGIT), modulation of antigen presentation (e.g., epigenetic agents restoring HLA-I), and targeted therapy of co-mutated genes (e.g., STK11/KEAP1) [93,94,95,96,97].
Despite this progress, many of these strategies remain in early-phase trials and lack definitive clinical validation. Future research must prioritise predictive biomarkers of resistance, longitudinal monitoring of immune phenotypes, and rational combinations tailored to the molecular and immunologic profile of each patient’s tumor.

4.1. Lymphocyte Activation Gene 3

LAG-3 (Lymphocyte-activation gene 3) is a surface protein expressed on effector T cells and regulatory T cells (Tregs), playing a key role in modulating the adaptive immune response. It is encoded by the LAG3 gene, which is localized on the short arm of chromosome 12 [98]. The interaction between LAG3 and its ligands (Major Histocompatibility Complex II (MHC-II) or Fibrinogen-like protein 1 (FGL1)) leads to reduce T cell function and cytokines production (inhibiting T cell receptor (TCR) signaling), contributing to the maintenance of immune homeostasis (Figure 2) [99]. Recently, LAG-3 has been identified as an important immune checkpoint in different cancer types, and its blockade has been associated with enhanced antitumor responses.
Although anti-LAG3 monotherapy has generally demonstrated limited antitumor efficacy, the combination of anti-LAG3 and anti-PD-1 has demonstrated synergistic activity in tumor models, leading to the development of the first anti-LAG3 mAbs, relatlimab [100].
Relatlimab is a humanized IgG4 mAb targeting LAG-3. In the RELATIVITY-047 trial, the combination of relatlimab plus nivolumab showed for the first time a significant improvement in PFS compared to nivolumab monotherapy in metastatic melanoma (10.1 months compared with 4.6 months, respectively) [101]. These findings led to the FDA approval of relatlimab in March 2022, marking it as the first anti-LAG3 therapy approved for use in metastatic melanoma [68].
Based on this rationale, relatlimab combined with nivolumab as neoadjuvant versus nivolumab monotherapy is under evaluation to treat resectable NSCLC patients, in the NEOpredict-Lung trial. The study showed a partial response rate of 27% in the combination arm versus 10% in nivolumab alone in patients with a positive expression of PD-L1 [67].
Alternative approaches, such as eftilagimod alpha, a soluble LAG-3 protein activating antigen-presenting cells (APCs), have shown promise in combination with pembrolizumab, particularly in PD-L1-high NSCLC patients, with 8.3% ORR and 33.3% Disease Control Rate (DCR) [102]. As LAG-3 inhibitors, such as relatlimab and eftilagimod, continue to reshape therapeutic paradigms, their integration with other immune-modulating agents holds great potential to advance the treatment of cancers.
Recently, several ongoing studies have identified other LAG-3 inhibitors (favezelimab, fianlimab, GLS-012, HLX26, and LBL-007), which have been tested as monotherapy or in combination with other ICIs in advanced solid tumors (Table 2). Although some of these agents exhibited good tolerability, they failed to demonstrate significant efficacy, especially in heavily pretreated or ICI-resistant populations [103]. These results highlight the challenges in effectively targeting the LAG-3 pathway within the complex immunosuppressive environment of NSCLC. Consequently, there is a pressing need to develop innovative strategies to inhibit LAG-3, including optimizing combination regimens with other ICIs, exploring novel biomarkers to select responsive patients, and designing next-generation LAG-3-targeted agents to overcome resistance mechanisms.

4.2. T Cell Immunoglobulin and Mucin-Domain-Containing-3

The T cell immunoglobulin and mucin-domain-containing-3 (TIM-3) gene is located on the long arm of chromosome 5. It encodes for a membrane protein that acts as an immune checkpoint receptor [104]. The protein is expressed in various immune cells, including T cells, natural killer (NK) cells, dendritic cells, and monocytes [105]. TIM-3 plays a crucial role in maintaining immune tolerance and regulating the immune response by inhibiting the activation and proliferation of T cells [106]. It interacts with multiple ligands, such as galectin-9, phosphatidylserine, HMGB1, and CEACAM1, to modulate immune activity in physiological and pathological contexts (Figure 2). In cancer, TIM-3 expression is often upregulated in exhausted T cells, contributing to immune evasion by tumors [105]. As a result, TIM-3 has emerged as a promising therapeutic target, with ongoing research focusing on its blockade to restore T cell function and enhance antitumor immunity. Recent phase 1 clinical trials have highlighted the potential of TIM-3 blockade in the treatment of NSCLC, particularly in combination with PD-1/PD-L1 immune checkpoint inhibitors, to improve outcomes in different cancer types.
In this context, cobolimab, a mAb of the IgG4 subclass targeting TIM-3, demonstrated a favorable safety profile both as monotherapy and in combination with dostarlimab (an anti-PD-1 agent), although its initial antitumor activity in NSCLC patients was limited [107]. In particular, the phase 1 AMBER study evaluated the combination of cobolimab and dostarlimab in patients with advanced or metastatic NSCLC previously treated with anti-PD(L)-1 therapy (Table 1). The aim was to explore a potential strategy to overcome acquired resistance to immunotherapy. The results showed an ORR of 8.3%, while the disease control rate DCR was 21.4%, indicating limited but potentially meaningful clinical activity in a heavily pretreated population. The safety profile of the combination was manageable, with adverse events mostly mild to moderate in severity. These preliminary data suggest that adding TIM-3 blockade to PD-1 inhibition could represent a promising approach to enhance responses in NSCLC patients refractory to standard immunotherapy, supporting further investigation in selected cohorts or in combination with other agents [57].
Recently, another humanized IgG4 mAb designed to block TIM-3 developed by Novartis, sabatolimab, was tested in combination with spartalizumab (a humanized IgG4 mAb targeting PD-1) in patients with NSCLC who had previously received anti-PD-1/L1 therapies. The combined approach demonstrated limited efficacy, with only 35% of patients achieving a stable response, while the majority experienced disease progression [108]. Despite its tolerability in combination, the modest results highlighted the need for further optimization of TIM-3 blockade in NSCLC. These findings support further investigation into TIM-3 as a therapeutic target to overcome resistance to PD-1/PD-L1 inhibitors. Current research focuses on improving the therapeutic impact of sabatolimab and other new anti-TIM-3 mAbs, through improved combination strategies or biomarker-based patient selection, to better address the challenges of immune resistance in NSCLC (Table 3).

4.3. T Cell Immunoreceptor with Immunoglobulin and ITIM Domain

T cell immunoreceptor with immunoglobulin and ITIM domain (TIGIT) gene is located on the long arm of chromosome 3. It encodes for a protein that acts as an immune checkpoint receptor expressed on T cells and natural killer (NK) cells, playing a critical role in suppressing anti-tumor immune responses [109]. The binding between TIGIT and its ligands CD155 (PVR) and CD112 is involved in the reduction of CD226 and TCR signaling pathway, which are essential for the activation of effector T cells (Figure 2) [110]. In particular, TIGIT competes with CD226 for binding to ligands expressed on tumor cells and antigen-presenting cells (APCs). This competition is crucial for regulating the immune response, as CD226 binding to ligands promotes homodimer formation and the activation of T and NK cells. In contrast, TIGIT prevents this interaction and recruits inhibitory signals through the ITIM domain, reducing immune cell proliferation and effector function [110]. Moreover, the interaction of TIGIT with CD155 on APCs can induce polarization toward a tolerogenic phenotype, further contributing to immunosuppression in the TME [111].
In NSCLC, overexpression of PVR and increased TIGIT expression correlate with impaired T cell activation, promoting immune exhaustion and reduced cytotoxicity [112]. This imbalance suppresses antitumor immunity and facilitates immune evasion. Targeting this axis, particularly through TIGIT blockade, is a promising therapeutic strategy aimed at restoring T cell function and enhancing the efficacy of immunotherapy in NSCLC [112].
Recently, it was suggested that combining TIGIT blockade with PD-1/PD-L1 inhibitors could facilitate the differentiation of activated T cells into effector or memory T cells, thereby boosting anti-tumor immune responses [113]. Clinical benefit has been associated with CD226 expression in patients with NSCLC treated with the anti-PD-L1 mAbs atezolizumab [22]. Mechanistically, PD-1 suppresses the phosphorylation of CD226 and CD28 through its ITIM-containing intracellular domain (ICD) [114]. Concurrently, TIGIT limits CD226 co-stimulation by blocking its interaction with the shared ligands. To fully restore CD226 signaling and maximize immune anti-tumor activity, simultaneous inhibition of both TIGIT and PD-1 pathways is required, providing a solid mechanistic basis for dual targeting in clinical applications [115]. The preliminary efficacy of the combination therapy involving the novel anti-TIGIT mAb, tiragolumab, and atezolizumab was evaluated for chemotherapy-naive patients with PD-L1 positive metastatic NSCLC, in the CITYSCAPE trial (Table 1) [62]. The trial aimed to assess the potential synergistic effect of blocking both TIGIT and PD-L1 pathways to enhance immune responses against tumors. In 2022, the preliminary results of this trial showed promising effects, with the combination therapy leading to an improved ORR and PFS compared to atezolizumab alone (31.3% vs. 16.2% and 5.4 vs. 3.6 months, respectively) [62]. Specifically, the combination demonstrated a higher clinical benefit in patients with high PD-L1 expression, suggesting that targeting multiple immune checkpoints may overcome resistance mechanisms observed with single-agent immunotherapy. These findings highlight the potential of dual checkpoint inhibition as a strategy to improve outcomes in NSCLC, particularly in patients who may not respond optimally to current therapies.
Despite the initial promise of TIGIT blockade, recent Phase II/III results have been less encouraging. Notably, The Phase III SKYSCRAPER-01 trial evaluated the combination of the anti-TIGIT monoclonal antibody tiragolumab with the anti–PD-L1 mAb atezolizumab as first-line therapy for patients with PD-L1–high (≥50%) advanced or metastatic NSCLC [69]. Although the combination showed a numerical improvement in both PFS (median 7.0 vs. 5.6 months; HR = 0.78) and OS (median 23.1 vs. 16.9 months; HR = 0.87) compared with atezolizumab monotherapy, these differences did not reach statistical significance [69]. Similarly, subsequent analyses did not confirm the early efficacy signals observed in CITYSCAPE study. Indeed, in 2024 the Phase II/III SKYSCRAPER-06 trial failed to demonstrate a statistically significant improvement in OS or PFS with tiragolumab plus atezolizumab and chemotherapy compared to pembrolizumab in combination with chemotherapy in non-SqCC NSCLC patients [116]. These outcomes have tempered the initial enthusiasm for TIGIT-targeted immunotherapy, underscoring the complexity of checkpoint interactions and the need for refined biomarker-driven strategies to identify responders.
Recently, other newly developed anti-TIGIT mAbs including four humanized IgG1 mAb targeted TIGIT (domvanalimab, ociperlimab, rilvegotomig, and tiragolumab) are under evaluation in several phase I/II studies for NSCLC patients (Table 4). Early findings indicate that these mAbs in combination with PD-1 inhibitors achieve higher response rates compared to PD-1/PD-L1 inhibition alone, likely driven by synergistic mechanisms such as enhanced activation of NK cells and CD8+ tumor-infiltrating lymphocytes (TILs). However, further investigations are necessary to establish the optimal sequencing of these therapies and to identify patient subgroups that would benefit most from early integration of chemotherapy.
In summary, while the TIGIT pathway remains a biologically compelling target for immune modulation in NSCLC, its clinical validation is still evolving. Future research should focus on unravelling mechanisms of primary and adaptive resistance, optimizing combination regimens, and stratifying patients based on CD226 expression, PD-L1 levels, and tumor immune contexture to fully harness the therapeutic potential of TIGIT inhibition.

4.4. Novel Potential Immune Checkpoints

Recently, the exploration of novel immune checkpoints, such as IDO1, GITR, NKG2A, CD38, CD47, OX-40 and adenosine receptors, is gaining attention as potential targets to overcome resistance to conventional ICIs. Recent preclinical and clinical studies are actively evaluating novel agents directed against these checkpoints to enhance anti-tumor immunity [117]. For example, targeting CD47, a “don’t eat me” signal that prevents macrophage-mediated phagocytosis, has shown promise in preclinical models and early-phase trials [118]. In a Phase I clinical trial evaluating the IgG4 anti-CD47 antibody magrolimab in combination with chemotherapy, encouraging preliminary responses were observed in advanced NSCLC patients, suggesting enhanced tumor clearance [119]. Similarly, inhibition of NKG2A, an inhibitory receptor expressed on NK cells and cytotoxic T cells, with the IgG4 mAb monalizumab in combination with durvalumab demonstrated improved response rates in early-stage studies, particularly in PD-L1-positive NSCLC [120]. Additionally, targeting the adenosine pathway, which suppresses immune responses in the TME, with adenosine receptor antagonists (A2AR) have shown promise in combination with PD-1 inhibitors in early-phase clinical trials [121,122]. Although most of these novel strategies are still in their early development stages, preliminary data highlight their potential to provide effective therapeutic options and further broaden the scope of immunotherapy for NSCLC treatment. Larger clinical studies will be essential to confirm their efficacy and safety profiles, paving the way for the integration of these approaches into future treatment regimens.

5. Novel Approaches to NSCLC Immunotherapy

The use of ICIs has revolutionized the treatment of many cancer types, including NSCLC. However, their use is associated with irAEs, which result from immune system activation against healthy tissues [123]. IrAEs can affect various organs and range in severity from mild to potentially life-threatening [124]. Their severity ranges from mild (grade 1–2) to potentially life-threatening (grade 3–4), with reported incidence in NSCLC patients of up to 60–70% for any-grade irAEs and approximately 10–15% for severe irAEs requiring hospitalization or immunosuppressive therapy [125,126].
Early recognition and proactive management are essential to minimize complications while preserving the anti-tumor efficacy of ICIs. Guidelines recommend regular monitoring, patient education, and prompt intervention with corticosteroids or other immunosuppressive agents, such as infliximab, for high-grade events [127]. Importantly, several studies indicate that appropriate management of irAEs does not necessarily compromise therapeutic benefit [128]. In fact, in some cases, the occurrence of certain irAEs, such as dermatologic or endocrine events, has been correlated with improved clinical outcomes, suggesting a complex relationship between immune activation and tumor control [129]. However, some data indicate that adherence to these recommendations remains suboptimal, with less than half of irAEs managed according to established protocols [130]. This highlights the need for improved clinician education and standardized documentation. Innovative strategies—such as nurse-led active monitoring and multidisciplinary supportive care—are emerging as valuable tools to enable earlier intervention, enhance patient safety, and promote a more patient-centered approach to immunotherapy management.
Emerging next-generation immunotherapy approaches in NSCLC aim to expand the therapeutic potential of immune-based treatments beyond traditional ICIs. Novel strategies include therapeutic agents like bispecific mAbs, personalized cancer vaccines and adoptive T-cell therapies, including the chimeric antigen receptor (CAR-T) cells engineered to recognize tumor-specific antigens, are advancing with promising preclinical and early clinical results in NSCLC [131,132]. These emerging therapies, combined with existing ICIs or used in novel combinations, hold great promise for improving outcomes in patients with advanced NSCLC, particularly those who are refractory to current immunotherapies [133].

5.1. Adoptive T Cell Tranfer

Adoptive T-cell therapy (ACT) is an advanced and innovative approach in cancer immunotherapy, using the patient’s immune cells to more effectively fight malignancies. This process involves extracting T lymphocytes from the patient, enhancing T cells antitumor properties through laboratory modification or expansion, and reinfusing them in the same patient to specific target and eliminate cancer cells [134]. The ACT encompasses different techniques, such as employing tumor-infiltrating lymphocytes (TILs), engineering T cells to express chimeric antigen receptors (CAR-T cells), or modifying them with tumor-specific T-cell receptors (TCRs).
Tumor-infiltrating lymphocytes (TILs), one of the most classic forms of ACT, exploiting the intrinsic ability of T cells to recognize and attack tumor antigens present within the TME. Although this strategy has shown promising results in specific cancer types, such as melanoma, the complexity of NSCLC poses significant challenges [135,136]. Indeed, the pronounced antigenic heterogeneity of NSCLC presents a substantial obstacle to identifying a uniform therapeutic target. Furthermore, the highly immunosuppressive TME significantly impairs the effectiveness of T-cell responses [137]. To overcome these limitations, research has led to the development of more advanced technologies, such as genetically engineered T cells, including CAR-T cells and TCR-engineered T cells, offering new perspective in the treatment of NSCLC. In particular, advancements in gene recombination techniques have led to the development of “super-lymphocytes” genetically engineered to enhance their specificity toward antigens overexpressed in tumors, even in the absence of natural reinforcement by APCs [138]. Specifically, T lymphocytes from the patient are extracted and undergo genetic engineering to express artificial antigen receptors known as “Chimeric Antigen Receptors” (CARs) (Figure 3). The receptor is composed of a single-chain variable fragment (scFv) derived from an antibody, which recognizes a specific tumor-associated antigen, fused with intracellular signaling domains that activate T-cell responses [139]. CARs are designed to bypass the requirement for antigen presentation by MHC molecules, allowing T cells to target tumor cells directly. Additionally, the receptor typically includes a costimulatory domain, such as CD28 or 4-1BB, to enhance the persistence and efficacy of the T cells [140].
The highly innovative CAR-T technology was initially developed by the University of Pennsylvania [141,142]. It has demonstrated remarkable success in hematologic malignancies and is being actively investigated for its potential for solid tumors including NSCLC [143]. In this regard, the application of CAR-T cell therapy in NSCLC has shown promising preliminary results in terms of safety, but its clinical efficacy remains limited. Several phase I and I/II trials have confirmed the feasibility and tolerability of this approach, reporting a low incidence of severe adverse events and only rare cases of cytokine release syndrome or pulmonary toxicity, which were generally manageable with corticosteroids. In the trial NCT01869166 on EGFR-CAR-T cells, 11 patients with metastatic EGFR-positive NSCLC received a single CAR-T infusion after lymphodepletion: no treatment-related serious AEs were observed, but only two partial responses and five cases of stable disease were reported, with a median PFS of 7 weeks, indicating transient antitumor activity [144].
From an engineering standpoint, innovations have sought to overcome the biological barriers typical of solid tumors. The use of non-viral systems (such as piggyBac) has streamlined manufacturing. In particular, the phase I clinical trial NCT03182816 evaluated the safety and feasibility of piggyBac-generated EGFR-targeted CAR-T cells in patients with advanced, relapsed/refractory EGFR-positive NSCLC. The therapy was well tolerated, with only mild to moderate fever (grade 1–3) as the most common AE and no severe cytokine release syndrome (CRS) reported [145]. CAR-T cells were detectable in the peripheral blood of eight patients after infusion; one patient achieved a durable partial response lasting over 13 months, six had stable disease, and two experienced disease progression. The median PFS was 7.1 months, and the median OS was 15.6 months, suggesting a good tolerability and promising clinical activity [145]. These findings support further investigation of piggyBac-engineered CAR-T therapy as a simpler and more cost-effective alternative to viral vector-based CAR-T cell generation in NSCLC.
Emerging targets like CD276 and PD-L1 have also gained attention for their diagnostic and immunological relevance in NSCLC [146]. CAR-T cells designed to target PD-L1 have exhibited potent efficacy against tumors with both high and low PD-L1 expression. Innovative strategies, such as combining CAR-T therapy with localized irradiation or modifying CAR-T cells to express receptors like CXCR5 for enhanced tumor infiltration, are being explored to overcome the challenges posed by the NSCLC immunosuppressive TME and antigenic heterogeneity [147]. Despite these advances, CAR-T therapy for NSCLC is still in its early stages, requiring further research to address hurdles like tumor antigen escape, limited tumor infiltration, and AEs, before it can achieve widespread clinical application.
Recently, new generations of “armored CAR-T cells,” designed to secrete immunostimulatory cytokines (IL-12, IL-7, IL-15, CCL19) or incorporating second-generation costimulatory domains (CD28, 4-1BB), have shown improved tumor infiltration and persistence in preclinical models [148]. However, significant challenges remain, including poor trafficking into the tumor microenvironment, antigenic heterogeneity leading to immune escape, limited in vivo persistence, and potential on-target/off-tumor toxicity due to antigen expression in normal tissues.
Despite a high rate of clinical responses, CAR-T therapy is unfortunately accompanied by several limits. One major challenge is the high cost and complexity of manufacturing CAR-T cells, which requires personalized cell engineering for each patient. Another critical limitation is the potential for severe side effects, including CRS and immune effector cell-associated neurotoxicity syndrome (ICANS), which can be life-threatening and require intensive management [149]. Some patients may also develop a condition known as “hemophagocytic lymphohistiocytosis” (HLH) or “macrophage activation syndrome” (MAS). This is observed in approximately 3.5% of patients treated with CAR-T cells and manifests with fever, hepatosplenomegaly, impaired liver function, and low blood cell counts [150]. Moreover, tumor relapse due to antigen escape, where cancer cells lose or downregulate the targeted antigen, remains a significant hurdle [151]. These challenges highlight the need for continued research to refine CAR-T cell engineering, improve their safety and efficacy, and expand their utility to a wider range of cancers.
The development of ACT targeting NSCLC poses substantial challenges but also significant opportunities for innovation. Key considerations include the identification of optimal tumor-specific antigens (TSAs) or tumor-associated antigens (TAAs) with minimal expression in normal tissues, mitigating off-tumor effects, and overcoming immune tolerance within the TME [152]. Addressing critical issues such as CAR-T cell-associated toxicities, antigen escape, heterogeneity, limited proliferation, and insufficient tumor infiltration requires cutting-edge synthetic biology approaches [153]. Innovations such as multispecific CARs targeting multiple antigens or different cell types (e.g., CD4, CD8, NK cells), spatiotemporally controllable CAR activation using switchable CARs (induced via ultrasound, light, drugs, or adapters), and safety mechanisms, like inducible caspase-9 switches, offer promising solutions [154]. Moreover, self-regulating and self-destructive CAR architectures are being designed to enhance safety, while multi-target CARs aim to address antigen heterogeneity in LC effectively [155]. Another promising avenue involves creating CAR-T cells tailored to specific LC genotypes, leveraging artificial intelligence and CRISPR technology to design synthetic receptors, such as SynNotch systems, that integrate chemical recognition events [156,157]. Researchers are also exploring the mass production of “off-the-shelf” CAR-T cells, potentially simplifying and reducing the costs of CAR-T therapy. The identification of neoantigens and the development of high-throughput screening tools are critical for optimizing CAR-T components and advancing the therapeutic potential of ACT for NSCLC (Table 5).
In summary, clinical and preclinical data indicate that CAR-T cell therapy in NSCLC represents a promising but still experimental platform, where therapeutic benefits are often transient. The future success of this strategy will depend on the development of more sophisticated constructs, combinatorial approaches with ICIs, oncolytic vaccines, or bispecific antibodies, and the identification of predictive biomarkers to better select patients most likely to respond ultimately aiming to enhance the durability and depth of antitumor responses in refractory lung cancer.

5.2. Oncological Vaccines

Oncological vaccines represent one of the most promising and innovative frontiers in the fight against cancer, offering new hope, particularly in the treatment of NSCLC. These vaccines are designed to enhance the patient immune system, directing it to selectively recognize and destroy cancer cells, sparing the healthy tissues [158].
Innovative approaches, such as multi-target vaccines that simultaneously target multiple tumor antigens, are showing significant potential. Combinations with immunomodulatory therapies, such as immune checkpoint inhibitors, further enhance treatment efficacy, creating a synergy that could overcome many current limitations [159].
Examples of oncological vaccines already in advanced stages of study include CIMAvax-EGF and TG4010.
The CIMAvax-EGF is a therapeutic vaccine developed in Cuba, designed to block the action of the epidermal growth factor (EGF), a protein that plays a critical role in the growth and proliferation of cancer cells [160]. CIMAvax-EGF aims to stimulate the patient immune system to produce antibodies against EGF, thereby reducing the stimulation of tumor growth. The distinctive feature of this vaccine is its ability to interfere with the binding of EGF to its receptor on the surface of cancer cells, preventing their proliferation [161]. CIMAvax-EGF has been approved in certain countries, such as Cuba, and is used in combination with other treatments, such as chemotherapy, to improve outcomes in patients with advanced NSCLC. Clinical studies have shown that treatment with CIMAvax-EGF can prolong OS, although its efficacy varies depending on the individual characteristics of the tumor and of the patient [162].
The TG4010 is another promising oncological vaccine, developed by the French biotechnology company Transgene [163]. This vaccine is a vector-based on the vaccinia virus, genetically modified to express the MUC1 antigen, a glycoprotein overexpressed on the surface of cancer cells, particularly in NSCLC [164]. TG4010 is designed to stimulate a targeted immune response against cancer cells expressing MUC1. The vaccine is often administered in combination with chemotherapy to enhance the effectiveness of the immune response and reduce tumor size. Clinical trials have demonstrated that TG4010 is well tolerated by patients and that its combination with chemotherapy can improve treatment response and extend survival [165]. Although the results are promising, further studies are required to optimize its clinical application and determine its long-term impact on survival and patients’ quality of life.
Recently, cutting-edge technologies, including the use of genetically modified dendritic cells (DCs) to more effectively activate the immune system, represent a crucial step forward in personalized therapy [166]. A significant milestone in this area is Sipuleucel-T, a DC-based vaccine that received the FDA approval for the treatment of metastatic prostate cancer, in 2010 [167]. Since then, DC vaccines have been the focus of numerous clinical studies. A promising approach involves the intratumoral administration of autologous DC vaccines, such as those modified with the CCL21 gene, to enhance the infiltration of T cells into tumors [168]. This has been demonstrated in studies involving patients with NSCLC. In particular, a Phase I study using AdCCL21-DC showed significant improvements in the activation of CD8+ T cells and the tumor-specific immune response [168,169]. Additionally, combining DC vaccines with immunotherapies, such as pembrolizumab has generated growing interest, as highlighted by ongoing studies.
Moreover, several innovative cancer vaccine strategies are under investigation in NSCLC, aiming to enhance anti-tumor immune responses in various clinical settings. In particular, PDClung01 is a therapeutic vaccine composed of irradiated plasmacytoid DC loaded with multiple HLA-A02:01-restricted tumor-associated peptides (including NY-ESO-1, MAGE-A3/A4, MUC1, survivin, and Melan-A) [170]. In a phase I/II study, patients with stage IV NSCLC (PD-L1 ≥ 50%) treated with high-dose PDClung01 plus pembrolizumab achieved an impressive ORR of 63.2% and a median PFS of 10.9 months, with acceptable safety [170].
Another promising candidate, CAN-2409, is an adenoviral vector carrying HSV-thymidine kinase that, combined with valacyclovir and ICI therapy, acts as an in situ vaccine. In a phase II trial of ICI-resistant NSCLC patients, the combination yielded a median OS of 22 months, with 65% of patients experiencing tumor shrinkage and no dose-limiting toxicities [171].
Additionally, BNT116, an RNA-lipoplex vaccine encoding six tumor antigens, showed an ORR of 35% and a disease control rate of 85% when combined with docetaxel in ICI- and chemotherapy-pretreated NSCLC patients [172]. Similarly, BI1361849, another RNA-based vaccine, was evaluated in combination with durvalumab with or without tremelimumab. While both regimens were well tolerated, the addition of tremelimumab did not improve efficacy, with arm A showing better outcomes (ORR 29%, mPFS 5.7 months) compared to arm B (ORR 11%, mPFS 2.5 months) [173].
Collectively, these studies highlight the potential of cancer vaccines as adjunctive strategies in NSCLC, particularly in overcoming resistance to ICIs. However, despite these promising results, significant challenges remain, including immune tolerance, the limited duration of the immune response, and the complexity of producing DC vaccines. Ongoing research is investigating innovative approaches, including next-generation DC vaccines, the use of different DC types such as plasmacytoid (pDC), conventional (cDC1), and DC-derived exosomes to further improve the efficacy and durability of immune responses against cancer (Table 6) [174,175,176].

5.3. Bispecific Antibodies

In recent years, bispecific antibodies (bsAbs) represent a promising innovation in the treatment of NSCLC, particularly in the context of immunotherapy [177]. These antibodies are designed to simultaneously recognize two different molecular targets, enabling a dual modulation of the TME [178]. In the case of NSCLC, the focus is primarily on immune checkpoints such as PD-1/PD-L1 and CTLA-4, which are known to play a crucial role in tumor immune evasion. BsAbs like amivantanab, cadonilimab, and QL1706, which simultaneously target PD-1 and CTLA-4, have shown promising preclinical and clinical results.
For instance, cadonilimab enhances tumor-specific T-cell activation by concurrently blocking these checkpoints while reducing treatment-related toxicity [179]. Clinical studies such as COMPASSION-01 and COMPASSION-03 have explored the efficacy and safety of cadonilimab in patients with advanced solid tumors, including those resistant to chemotherapy and immunotherapy, demonstrating objective responses and acceptable tolerability [180,181]. However, in the specific context of NSCLC, the results have been more variable, with limited objective responses in subgroups of patients with primary or acquired resistance to PD-1/PD-L1 inhibitors.
Another relevant bsAb, QL1706, which combines an anti-PD-1 antibody with an anti-CTLA-4 antibody, has shown anti-tumor activity in advanced solid tumors, including NSCLC, with an improved tolerability profile due to pharmacokinetic optimization and reduced irAEs [182].
Recent clinical data underscore the growing role of amivantamab, a bsAb targeting both EGFR and MET, in the treatment of NSCLC, particularly among patients with EGFR mutations and MET alterations [183]. MET amplification has emerged as a key mechanism of resistance to osimertinib, both in treatment-naïve patients and those with acquired T790M mutations, as well as in cases with de novo MET amplification, supporting the strategy of co-targeting MET early in the treatment course [184]. Amivantamab, developed via the DuoBody platform, is the first and only bsAb approved for NSCLC and has shown significant clinical activity across several patient populations [177]. In the phase 3 PAPILLON trial, the combination of amivantamab and chemotherapy in patients with EGFR exon 20 insertions yielded superior outcomes in response rate (73%) and PFS (11.4 months) compared to chemotherapy alone (6.7 months), leading to its FDA approval as a first-line treatment in this setting [185].
Furthermore, the MARIPOSA phase 3 trial demonstrated that combining lazertinib with amivantamab in treatment-naïve patients with common EGFR mutations significantly improved PFS (23.7 months vs. 16.6 months with osimertinib) [186,187]. This combination has received FDA approval for first-line treatment in patients with EGFR exon 19 deletions or exon 21 L858R mutations. Notably, subgroup analyses revealed consistent PFS benefits among high-risk populations, including those with brain or liver metastases, TP53 co-mutations, and detectable ctDNA. Evidence also suggests intracranial activity, including in patients with leptomeningeal disease [188].
Finally, in patients with uncommon EGFR mutations, the CHRYSALIS-2 study showed encouraging efficacy with lazertinib and amivantamab, both in treatment-naïve and pretreated populations, with ORRs ranging from 48% to 57% and PFS from 7.8 to 19.5 months [189].
Simultaneously, bsAbs combining immunotherapeutic targeting with other pathways are opening new therapeutic avenues.
In this context, ivonescimab (anti-PD-1 and VEGF), combined with chemotherapy, has demonstrated significant anti-tumor activity as a first-line therapy in advanced NSCLC without driver mutations or in patients resistant to prior treatments [190,191]. In particular, in the Phase III HARMONi-2 trial, ivonescimab monotherapy demonstrated a median PFS of 11.1 months, compared to 5.8 months with pembrolizumab, representing a 49% reduction in the risk of disease progression or death (HR = 0.51; p < 0.0001) (Table 1). These benefits were consistent across various subgroups, including patients with both low (1–49%) and high (≥50%) PD-L1 expression, as well as those with squamous and non-squamous histology [63]. Regarding toxicity, Ivonescimab displayed a manageable safety profile, with grade ≥ 3 treatment-related adverse events occurring in 29% of patients, the most frequent being hypertension (5%). Serious treatment-related adverse events were reported in 21% of cases [63]. Based on these encouraging outcomes, ivonescimab has been approved in China in combination with chemotherapy for the treatment of EGFR-TKI-resistant SqCC and non-SqCC NSCLC, and is currently under priority review as a first-line monotherapy for PD-L1-positive NSCLC [192]. These findings underscore the potential of bsAbs not only to enhance immune responses but also to overcome resistance limiting the effectiveness of traditional monotherapies.
A new frontier, still under investigation, could be represented by new generations of bsAbs, including the Bispecific T-Cell Engagers (BiTEs) and the Bispecific Nanobodies (BsNb). These two types of bsAbs have been emerging as promising strategies for the treatment of NSCLC. In particular, BiTEs are designed to recruit and activate cytotoxic T cells by simultaneously binding to a tumor antigen, such as EGFR or MET, and the CD3 receptor on T cells, facilitating targeted activation and tumor cell lysis [193]. On the other hand, Bispecific Nanobodies, derived from single-domain antibodies, offer additional advantages due to their smaller size, which allows for better tumor penetration and faster systemic clearance, improving their safety profile [194]. These new approaches are particularly promising in the immunosuppressive microenvironment of NSCLC, where they can overcome resistance to ICIs and amplify the antitumor response.
Despite these advances, significant challenges remain, such as identifying predictive biomarkers of response and managing associated toxicity. Ongoing clinical trials, such as AK104, SSGJ-707, and MCLA-128 are helping to better define the role of these agents in NSCLC therapy and establish new therapeutic paradigms (Table 7). If confirmed, bsAbs could represent a milestone in NSCLC immunotherapy, offering more effective and personalized options for patients.

6. Conclusions

The development of advanced diagnostic tests with greater specificity in identifying and characterizing oncological diseases has facilitated a shift from a traditional, generalized clinical model to a personalized diagnostic and therapeutic approach. This evolution enables tailored patient care, providing increasingly accurate and effective therapeutic strategies. In this context, molecular profiling plays a fundamental role, offering crucial information not only at diagnosis but also throughout disease evolution, thus supporting dynamic and evidence-based treatment decisions. This approach represents a model to follow, because it will provide the patient with greater possibilities of receiving increasingly effective therapies, minimizing adverse events due to the administration of non-targeted therapies, with consequent undoubted clinical benefits for the patient.
The past ten years have witnessed the development of the immunotherapy based on specific mAbs, the ICIs. When ICIs target the immune-checkpoint of the T cell reaction, the host immune system is reactivated and better able to identify and eliminate NSCLC cells. The ICIs have been demonstrated to be successful in lowering tumor burden and enhancing the prognosis of LC patients, either alone or in combination with other ICIs, conventional chemotherapy. However, one of the main disadvantages is the resistance that some patients develop to these therapies, reducing long-term efficacy. Recently, there has been growing interest in clinical trials, particularly focusing on innovative immunotherapeutic approaches. These include the use of new immune checkpoint inhibitors targeting different pathways to avoid resistance, and CAR-T cell therapy, which could potentially offer more targeted and personalized responses. Additionally, oncological vaccines and bispecific antibodies are the subject of intense studies, as they may stimulate the immune system to recognize specific tumor antigens, providing new therapeutic options for patients who do not respond to conventional therapies. However, although preliminary data from early-phase trials have shown encouraging signals of efficacy, several late-phase studies have yielded disappointing or inconclusive results, tempering initial optimism. These inconsistencies highlight the complexity of tumor–immune system interactions and underscore the need for more refined patient selection and biomarker-driven approaches. Finally, while immunotherapy continues to represent a cornerstone of NSCLC treatment, its future success will depend on overcoming resistance mechanisms and validating these novel approaches through robust clinical evidence.

Author Contributions

Conceptualization, G.M.L. and K.M.; writing—original draft preparation, G.M.L. and G.S.; writing—review and editing, G.S., P.F. and K.M.; visualization, G.M.L. and G.S.; supervision, P.F. and K.M.; project administration, P.F. and K.M.; funding acquisition, G.M.L., P.F., and K.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in this study are included in the article. Further inquiries can be directed at the corresponding authors.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global Cancer Statistics 2022: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef]
  2. Gridelli, C.; Rossi, A.; Carbone, D.P.; Guarize, J.; Karachaliou, N.; Mok, T.; Petrella, F.; Spaggiari, L.; Rosell, R. Non-Small-Cell Lung Cancer. Nat. Rev. Dis. Prim. 2015, 1, 15009. [Google Scholar] [CrossRef]
  3. Jemal, A.; Ma, J.; Rosenberg, P.S.; Siegel, R.; Anderson, W.F. Increasing Lung Cancer Death Rates Among Young Women in Southern and Midwestern States. J. Clin. Oncol. 2012, 30, 2739–2744. [Google Scholar] [CrossRef]
  4. Haddadin, S.; Perry, M.C. History of Small-Cell Lung Cancer. Clin. Lung Cancer 2011, 12, 87–93. [Google Scholar] [CrossRef]
  5. de Sousa, V.M.L.; Carvalho, L. Heterogeneity in Lung Cancer. Pathobiology 2018, 85, 96–107. [Google Scholar] [CrossRef]
  6. Travis, W.D.; Brambilla, E.; Nicholson, A.G.; Yatabe, Y.; Austin, J.H.M.; Beasley, M.B.; Chirieac, L.R.; Dacic, S.; Duhig, E.; Flieder, D.B.; et al. The 2015 World Health Organization classification of lung tumors: Impact of genetic, clinical and radiologic advances since the 2004 classification. J. Thorac. Oncol. 2015, 10, 1243–1260. [Google Scholar] [CrossRef]
  7. Travis, W.D.; Brambilla, E.; Riely, G.J. New Pathologic Classification of Lung Cancer: Relevance for Clinical Practice and Clinical Trials. J. Clin. Oncol. 2013, 31, 992–1001. [Google Scholar] [CrossRef]
  8. Petersen, I. The Morphological and Molecular Diagnosis of Lung Cancer. Dtsch. Aerzteblatt Online 2011, 108, 525–531. [Google Scholar] [CrossRef]
  9. Travis, W.D.; Brambilla, E.; Noguchi, M.; Nicholson, A.G.; Geisinger, K.R.; Yatabe, Y.; Beer, D.G.; Powell, C.A.; Riely, G.J.; Van Schil, P.E.; et al. International Association for the Study of Lung Cancer/American Thoracic Society/European Respiratory Society International Multidisciplinary Classification of Lung Adenocarcinoma. J. Thorac. Oncol. 2011, 6, 244–285. [Google Scholar] [CrossRef]
  10. Travis, W.D.; Brambilla, E.; Burke, A.P.; Marx, A.; Nicholson, A.G. Introduction to The 2015 World Health Organization Classification of Tumors of the Lung, Pleura, Thymus, and Heart. J. Thorac. Oncol. 2015, 10, 1240–1242. [Google Scholar] [CrossRef]
  11. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer Statistics, 2019. CA Cancer J. Clin. 2019, 69, 7–34. [Google Scholar] [CrossRef]
  12. Leone, G.M.; Candido, S.; Lavoro, A.; Vivarelli, S.; Gattuso, G.; Calina, D.; Libra, M.; Falzone, L. Clinical Relevance of Targeted Therapy and Immune-Checkpoint Inhibition in Lung Cancer. Pharmaceutics 2023, 15, 1252. [Google Scholar] [CrossRef]
  13. Shah, A.; Apple, J.; Belli, A.J.; Barcellos, A.; Hansen, E.; Fernandes, L.L.; Zettler, C.M.; Wang, C.-K. Real-World Study of Disease-Free Survival & Patient Characteristics Associated with Disease-Free Survival in Early-Stage Non-Small Cell Lung Cancer: A Retrospective Observational Study. Cancer Treat. Res. Commun. 2023, 36, 100742. [Google Scholar] [CrossRef]
  14. Jeon, D.S.; Kim, H.C.; Kim, S.H.; Kim, T.-J.; Kim, H.K.; Moon, M.H.; Beck, K.S.; Suh, Y.-G.; Song, C.; Ahn, J.S.; et al. Five-Year Overall Survival and Prognostic Factors in Patients with Lung Cancer: Results from the Korean Association of Lung Cancer Registry (KALC-R) 2015. Cancer Res. Treat. 2023, 55, 103–111. [Google Scholar] [CrossRef]
  15. Athey, V.L.; Walters, S.J.; Rogers, T.K. Symptoms at Lung Cancer Diagnosis Are Associated with Major Differences in Prognosis. Thorax 2018, 73, 1177–1181. [Google Scholar] [CrossRef]
  16. Ganti, A.K.; Klein, A.B.; Cotarla, I.; Seal, B.; Chou, E. Update of Incidence, Prevalence, Survival, and Initial Treatment in Patients with Non–Small Cell Lung Cancer in the US. JAMA Oncol. 2021, 7, 1824. [Google Scholar] [CrossRef]
  17. Asmara, O.D.; Hardavella, G.; Ramella, S.; Petersen, R.H.; Tietzova, I.; Boerma, E.C.; Tenda, E.D.; Bouterfas, A.; Heuvelmans, M.A.; van Geffen, W.H. Stage III NSCLC Treatment Options: Too Many Choices. Breathe 2024, 20, 240047. [Google Scholar] [CrossRef]
  18. Park, S.; Ock, C.-Y.; Kim, H.; Pereira, S.; Park, S.; Ma, M.; Choi, S.; Kim, S.; Shin, S.; Aum, B.J.; et al. Artificial Intelligence–Powered Spatial Analysis of Tumor-Infiltrating Lymphocytes as Complementary Biomarker for Immune Checkpoint Inhibition in Non–Small-Cell Lung Cancer. J. Clin. Oncol. 2022, 40, 1916–1928. [Google Scholar] [CrossRef]
  19. Federico, L.; McGrail, D.J.; Bentebibel, S.-E.; Haymaker, C.; Ravelli, A.; Forget, M.-A.; Karpinets, T.; Jiang, P.; Reuben, A.; Negrao, M.V.; et al. Distinct Tumor-Infiltrating Lymphocyte Landscapes Are Associated with Clinical Outcomes in Localized Non-Small-Cell Lung Cancer. Ann. Oncol. 2022, 33, 42–56. [Google Scholar] [CrossRef]
  20. Kunimasa, K.; Goto, T. Immunosurveillance and Immunoediting of Lung Cancer: Current Perspectives and Challenges. Int. J. Mol. Sci. 2020, 21, 597. [Google Scholar] [CrossRef]
  21. Qiang, M.; Liu, H.; Yang, L.; Wang, H.; Guo, R. Immunotherapy for Small Cell Lung Cancer: The Current State and Future Trajectories. Discov. Oncol. 2024, 15, 355. [Google Scholar] [CrossRef]
  22. Desai, A.; Peters, S. Immunotherapy-Based Combinations in Metastatic NSCLC. Cancer Treat. Rev. 2023, 116, 102545. [Google Scholar] [CrossRef]
  23. Cascone, T.; Fradette, J.; Pradhan, M.; Gibbons, D.L. Tumor Immunology and Immunotherapy of Non-Small-Cell Lung Cancer. Cold Spring Harb. Perspect. Med. 2022, 12, a037895. [Google Scholar] [CrossRef]
  24. Vaddepally, R.K.; Kharel, P.; Pandey, R.; Garje, R.; Chandra, A.B. Review of Indications of FDA-Approved Immune Checkpoint Inhibitors per NCCN Guidelines with the Level of Evidence. Cancers 2020, 12, 738. [Google Scholar] [CrossRef] [PubMed]
  25. Fehrenbacher, L.; Spira, A.; Ballinger, M.; Kowanetz, M.; Vansteenkiste, J.; Mazieres, J.; Park, K.; Smith, D.; Artal-Cortes, A.; Lewanski, C.; et al. Atezolizumab versus Docetaxel for Patients with Previously Treated Non-Small-Cell Lung Cancer (POPLAR): A Multicentre, Open-Label, Phase 2 Randomised Controlled Trial. Lancet 2016, 387, 1837–1846, Erratum in Lancet 2017, 389, e5. [Google Scholar] [CrossRef]
  26. 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]
  27. 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] [PubMed]
  28. Zhou, F.; Qiao, M.; Zhou, C. The Cutting-Edge Progress of Immune-Checkpoint Blockade in Lung Cancer. Cell. Mol. Immunol. 2021, 18, 279–293. [Google Scholar] [CrossRef]
  29. Dong, Y.; Sun, Q.; Zhang, X. PD-1 and Its Ligands Are Important Immune Checkpoints in Cancer. Oncotarget 2017, 8, 2171–2186. [Google Scholar] [CrossRef]
  30. Shen, X.; Zhang, L.; Li, J.; Li, Y.; Wang, Y.; Xu, Z.-X. Recent Findings in the Regulation of Programmed Death Ligand 1 Expression. Front. Immunol. 2019, 10, 1337. [Google Scholar] [CrossRef]
  31. Pabla, S.; Conroy, J.M.; Nesline, M.K.; Glenn, S.T.; Papanicolau-Sengos, A.; Burgher, B.; Hagen, J.; Giamo, V.; Andreas, J.; Lenzo, F.L.; et al. Proliferative Potential and Resistance to Immune Checkpoint Blockade in Lung Cancer Patients. J. Immunother. Cancer 2019, 7, 27. [Google Scholar] [CrossRef]
  32. Mariniello, A.; Borgeaud, M.; Weiner, M.; Frisone, D.; Kim, F.; Addeo, A. Primary and Acquired Resistance to Immunotherapy with Checkpoint Inhibitors in NSCLC: From Bedside to Bench and Back. BioDrugs 2025, 39, 215–235. [Google Scholar] [CrossRef]
  33. Arulraj, T.; Wang, H.; Deshpande, A.; Varadhan, R.; Emens, L.A.; Jaffee, E.M.; Fertig, E.J.; Santa-Maria, C.A.; Popel, A.S. Virtual Patient Analysis Identifies Strategies to Improve the Performance of Predictive Biomarkers for PD-1 Blockade. Proc. Natl. Acad. Sci. USA 2024, 121, e2410911121. [Google Scholar] [CrossRef]
  34. Sha, D.; Jin, Z.; Budczies, J.; Kluck, K.; Stenzinger, A.; Sinicrope, F.A. Tumor Mutational Burden as a Predictive Biomarker in Solid Tumors. Cancer Discov. 2020, 10, 1808–1825. [Google Scholar] [CrossRef] [PubMed]
  35. Vryza, P.; Fischer, T.; Mistakidi, E.; Zaravinos, A. Tumor Mutation Burden in the Prognosis and Response of Lung Cancer Patients to Immune-Checkpoint Inhibition Therapies. Transl. Oncol. 2023, 38, 101788. [Google Scholar] [CrossRef]
  36. Ben Dori, S.; Aizic, A.; Sabo, E.; Hershkovitz, D. Spatial Heterogeneity of PD-L1 Expression and the Risk for Misclassification of PD-L1 Immunohistochemistry in Non-Small Cell Lung Cancer. Lung Cancer 2020, 147, 91–98. [Google Scholar] [CrossRef] [PubMed]
  37. Kim, S.Y.; Kim, T.-E.; Park, C.K.; Yoon, H.-K.; Sa, Y.J.; Kim, H.R.; Woo, I.S.; Kim, T.-J. Comprehensive Comparison of 22C3 and SP263 PD-L1 Expression in Non-Small-Cell Lung Cancer Using Routine Clinical and Conditioned Archives. Cancers 2022, 14, 3138. [Google Scholar] [CrossRef]
  38. Ahmed, J.; Das, B.; Shin, S.; Chen, A. Challenges and Future Directions in the Management of Tumor Mutational Burden-High (TMB-H) Advanced Solid Malignancies. Cancers 2023, 15, 5841. [Google Scholar] [CrossRef]
  39. Zgura, A.; Chipuc, S.; Bacalbasa, N.; Haineala, B.; Rodica, A.; Sebastian, V. Evaluating Tumour Mutational Burden as a Key Biomarker in Personalized Cancer Immunotherapy: A Pan-Cancer Systematic Review. Cancers 2025, 17, 480. [Google Scholar] [CrossRef] [PubMed]
  40. McGrail, D.J.; Pilié, P.G.; Rashid, N.U.; Voorwerk, L.; Slagter, M.; Kok, M.; Jonasch, E.; Khasraw, M.; Heimberger, A.B.; Lim, B.; et al. High Tumor Mutation Burden Fails to Predict Immune Checkpoint Blockade Response across All Cancer Types. Ann. Oncol. 2021, 32, 661–672. [Google Scholar] [CrossRef]
  41. Thagaard, J.; Broeckx, G.; Page, D.B.; Jahangir, C.A.; Verbandt, S.; Kos, Z.; Gupta, R.; Khiroya, R.; Abduljabbar, K.; Acosta Haab, G.; et al. Pitfalls in Machine Learning-based Assessment of Tumor-infiltrating Lymphocytes in Breast Cancer: A Report of the International Immuno-Oncology Biomarker Working Group on Breast Cancer. J. Pathol. 2023, 260, 498–513. [Google Scholar] [CrossRef] [PubMed]
  42. Thommen, D.S.; Schumacher, T.N. T Cell Dysfunction in Cancer. Cancer Cell 2018, 33, 547–562. [Google Scholar] [CrossRef] [PubMed]
  43. Brahmer, J.; Reckamp, K.L.; Baas, P.; Crinò, L.; Eberhardt, W.E.E.; Poddubskaya, E.; Antonia, S.; Pluzanski, A.; Vokes, E.E.; Holgado, E.; et al. Nivolumab versus Docetaxel in Advanced Squamous-Cell Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2015, 373, 123–135. [Google Scholar] [CrossRef] [PubMed]
  44. Sul, J.; Blumenthal, G.M.; Jiang, X.; He, K.; Keegan, P.; Pazdur, R. FDA Approval Summary: Pembrolizumab for the Treatment of Patients with Metastatic Non-Small Cell Lung Cancer Whose Tumors Express Programmed Death-Ligand 1. Oncologist 2016, 21, 643–650. [Google Scholar] [CrossRef]
  45. 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]
  46. Rittmeyer, A.; Barlesi, F.; Waterkamp, D.; Park, K.; Ciardiello, F.; von Pawel, J.; Gadgeel, S.M.; Hida, T.; Kowalski, D.M.; Dols, M.C.; et al. Atezolizumab versus Docetaxel in Patients with Previously Treated Non-Small-Cell Lung Cancer (OAK): A Phase 3, Open-Label, Multicentre Randomised Controlled Trial. Lancet 2017, 389, 255–265. [Google Scholar] [CrossRef]
  47. Antonia, S.J.; Villegas, A.; Daniel, D.; Vicente, D.; Murakami, S.; Hui, R.; Kurata, T.; Chiappori, A.; Lee, K.H.; de Wit, M.; et al. Overall Survival with Durvalumab After Chemoradiotherapy in Stage III NSCLC. N. Engl. J. Med. 2018, 379, 2342–2350. [Google Scholar] [CrossRef]
  48. Faivre-Finn, C.; Vicente, D.; Kurata, T.; Planchard, D.; Paz-Ares, L.; Vansteenkiste, J.F.; Spigel, D.R.; Garassino, M.C.; Reck, M.; Senan, S.; et al. Four-Year Survival with Durvalumab After Chemoradiotherapy in Stage III NSCLC—An Update from the PACIFIC Trial. J. Thorac. Oncol. 2021, 16, 860–867. [Google Scholar] [CrossRef]
  49. 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]
  50. Sezer, A.; Kilickap, S.; Gümüş, M.; Bondarenko, I.; Özgüroğlu, M.; Gogishvili, M.; Turk, H.M.; Cicin, I.; Bentsion, D.; Gladkov, O.; et al. Cemiplimab Monotherapy for First-Line Treatment of Advanced Non-Small-Cell Lung Cancer with PD-L1 of at Least 50%: A Multicentre, Open-Label, Global, Phase 3, Randomised, Controlled Trial. Lancet 2021, 397, 592–604. [Google Scholar] [CrossRef]
  51. Özgüroğlu, M.; Kilickap, S.; Sezer, A.; Gümüş, M.; Bondarenko, I.; Gogishvili, M.; Nechaeva, M.; Schenker, M.; Cicin, I.; Ho, G.F.; et al. First-Line Cemiplimab Monotherapy and Continued Cemiplimab beyond Progression plus Chemotherapy for Advanced Non-Small-Cell Lung Cancer with PD-L1 50% or More (EMPOWER-Lung 1): 35-Month Follow-up from a Mutlicentre, Open-Label, Randomised, Phase 3 Trial. Lancet Oncol. 2023, 24, 989–1001, Erratum in Lancet Oncol. 2023, 24, e405. [Google Scholar] [CrossRef]
  52. Gogishvili, M.; Melkadze, T.; Makharadze, T.; Giorgadze, D.; Dvorkin, M.; Penkov, K.; Laktionov, K.; Nemsadze, G.; Nechaeva, M.; Rozhkova, I.; et al. Cemiplimab plus Chemotherapy versus Chemotherapy Alone in Non-Small Cell Lung Cancer: A Randomized, Controlled, Double-Blind Phase 3 Trial. Nat. Med. 2022, 28, 2374–2380. [Google Scholar] [CrossRef]
  53. Passaro, A.; Brahmer, J.; Antonia, S.; Mok, T.; Peters, S. Managing Resistance to Immune Checkpoint Inhibitors in Lung Cancer: Treatment and Novel Strategies. J. Clin. Oncol. 2022, 40, 598–610. [Google Scholar] [CrossRef]
  54. Wei, K.; Zhang, H.; Yang, S.; Cui, Y.; Zhang, B.; Liu, J.; Tang, L.; Tan, Y.; Liu, S.; Chen, S.; et al. Chemo-Drugs in Cell Microparticles Reset Antitumor Activity of Macrophages by Activating Lysosomal P450 and Nuclear HnRNPA2B1. Signal Transduct. Target. Ther. 2023, 8, 22. [Google Scholar] [CrossRef]
  55. Yatabe, Y. Molecular Pathology of Non-small Cell Carcinoma. Histopathology 2024, 84, 50–66. [Google Scholar] [CrossRef] [PubMed]
  56. Hu, A.; Sun, L.; Lin, H.; Liao, Y.; Yang, H.; Mao, Y. Harnessing Innate Immune Pathways for Therapeutic Advancement in Cancer. Signal Transduct. Target. Ther. 2024, 9, 68. [Google Scholar] [CrossRef] [PubMed]
  57. Davar, D.; Eroglu, Z.; Milhem, M.; Becerra, C.; Gutierrez, M.; Ribas, A.; Di Pace, B.; 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. In Proceedings of the Regular and Young Investigator Award Abstracts, San Diego, CA, USA, 1–5 November 2023; BMJ Publishing Group Ltd.: London, UK, 2023; p. A678. [Google Scholar]
  58. Reck, M.; Luft, A.; Szczesna, A.; Havel, L.; Kim, S.-W.; Akerley, W.; Pietanza, M.C.; Wu, Y.; Zielinski, C.; Thomas, M.; et al. Phase III Randomized Trial of Ipilimumab Plus Etoposide and Platinum Versus Placebo Plus Etoposide and Platinum in Extensive-Stage Small-Cell Lung Cancer. J. Clin. Oncol. 2016, 34, 3740–3748. [Google Scholar] [CrossRef]
  59. 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, Erratum in Lancet Oncol. 2021, 22, e92. [Google Scholar] [CrossRef]
  60. Carbone, D.P.; Ciuleanu, T.-E.; Cobo, M.; Schenker, M.; Zurawski, B.; Menezes, J.; Richardet, E.; Felip, E.; Cheng, Y.; Juan-Vidal, O.; et al. Nivolumab plus Ipilimumab with Chemotherapy as First-Line Treatment of Patients with Metastatic Non-Small-Cell Lung Cancer: Final, 6-Year Outcomes from CheckMate 9LA. ESMO Open 2025, 10, 105123. [Google Scholar] [CrossRef] [PubMed]
  61. 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]
  62. 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]
  63. Xiong, A.; Wang, L.; Chen, J.; Wu, L.; Liu, B.; Yao, J.; Zhong, H.; Li, J.; Cheng, Y.; Sun, Y.; et al. Ivonescimab versus Pembrolizumab for PD-L1-Positive Non-Small Cell Lung Cancer (HARMONi-2): A Randomised, Double-Blind, Phase 3 Study in China. Lancet 2025, 405, 839–849, Erratum in Lancet 2025, 406, 2062. [Google Scholar] [CrossRef]
  64. Nishio, M.; Barlesi, F.; West, H.; Ball, S.; Bordoni, R.; Cobo, M.; Longeras, P.D.; Goldschmidt, J.; Novello, S.; Orlandi, F.; et al. Atezolizumab Plus Chemotherapy for First-Line Treatment of Nonsquamous NSCLC: Results from the Randomized Phase 3 IMpower132 Trial. J. Thorac. Oncol. 2021, 16, 653–664. [Google Scholar] [CrossRef]
  65. 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] [PubMed]
  66. Paz-Ares, L.; Luft, A.; Vicente, D.; Tafreshi, A.; Gümüş, M.; Mazières, J.; Hermes, B.; Çay Şenler, F.; Csőszi, T.; Fülöp, A.; et al. Pembrolizumab plus Chemotherapy for Squamous Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 379, 2040–2051. [Google Scholar] [CrossRef]
  67. 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] [PubMed]
  68. Long, G.V.; Lipson, E.J.; Hodi, F.S.; Ascierto, P.A.; Larkin, J.; Lao, C.; Grob, J.-J.; Ejzykowicz, F.; Moshyk, A.; Garcia-Horton, V.; et al. First-Line Nivolumab Plus Relatlimab Versus Nivolumab Plus Ipilimumab in Advanced Melanoma: An Indirect Treatment Comparison Using RELATIVITY-047 and CheckMate 067 Trial Data. J. Clin. Oncol. 2024, 42, 3926–3934. [Google Scholar] [CrossRef] [PubMed]
  69. Peters, S.; Herbst, R.; Horinouchi, H.; Paz-Ares, L.; Johnson, M.; Solomon, B.; Gumus, M.; Erman, M.; Bondarenko, I.; Kim, D.-W.; et al. Abstract CT051: SKYSCRAPER-01: A Phase III, Randomized Trial of Tiragolumab (Tira) + Atezolizumab (Atezo) versus Placebo (Pbo) + Atezo in Patients (Pts) with Previously-Untreated PD-L1-High, Locally Advanced Unresectable/Metastatic NSCLC. Cancer Res. 2025, 85, CT051. [Google Scholar] [CrossRef]
  70. 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]
  71. Ibarrondo, F.J.; Comin-Anduix, B.; Escuin-Ordinas, H. Tremelimumab: Research and Clinical Development. Onco. Targets. Ther. 2016, 9, 1767. [Google Scholar] [CrossRef]
  72. Buchbinder, E.I.; Desai, A. CTLA-4 and PD-1 Pathways: Similarities, Differences, and Implications of Their Inhibition. Am. J. Clin. Oncol. 2016, 39, 98–106. [Google Scholar] [CrossRef]
  73. Lynch, T.J.; Bondarenko, I.; Luft, A.; Serwatowski, P.; Barlesi, F.; Chacko, R.; Sebastian, M.; Neal, J.; Lu, H.; Cuillerot, J.-M.; et al. Ipilimumab in Combination with Paclitaxel and Carboplatin As First-Line Treatment in Stage IIIB/IV Non–Small-Cell Lung Cancer: Results from a Randomized, Double-Blind, Multicenter Phase II Study. J. Clin. Oncol. 2012, 30, 2046–2054. [Google Scholar] [CrossRef]
  74. Lauriola, A.; Davalli, P.; Marverti, G.; Santi, S.; Caporali, A.; D’Arca, D. Targeting the Interplay of Independent Cellular Pathways and Immunity: A Challenge in Cancer Immunotherapy. Cancers 2023, 15, 3009. [Google Scholar] [CrossRef]
  75. Leisha, A.E. Chemotherapy and Tumor Immunity: An Unexpected Collaboration. Front. Biosci. 2008, 13, 249. [Google Scholar] [CrossRef]
  76. Hendriks, L.E.L.; Menis, J.; Remon, J. First-Line Immune-Chemotherapy Combination for Squamous NSCLC Is Already a Reality. Transl. Lung Cancer Res. 2020, 9, 819–823. [Google Scholar] [CrossRef]
  77. Zhang, D.; Zhao, J.; Zhang, Y.; Jiang, H.; Liu, D. Revisiting Immune Checkpoint Inhibitors: New Strategies to Enhance Efficacy and Reduce Toxicity. Front. Immunol. 2024, 15, 1490129. [Google Scholar] [CrossRef]
  78. Vellanki, P.J.; Mulkey, F.; Jaigirdar, A.A.; Rodriguez, L.; Wang, Y.; Xu, Y.; Zhao, H.; Liu, J.; Howe, G.; Wang, J.; et al. FDA Approval Summary: Nivolumab with Ipilimumab and Chemotherapy for Metastatic Non–Small Cell Lung Cancer, A Collaborative Project Orbis Review. Clin. Cancer Res. 2021, 27, 3522–3527. [Google Scholar] [CrossRef] [PubMed]
  79. 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]
  80. Peters, S.; Paz-Ares, L.G.; Reck, M.; Carbone, D.P.; Brahmer, J.R.; Borghaei, H.; Lu, S.; O’Byrne, K.J.; John, T.; Ciuleanu, T.-E.; et al. Long-Term Survival Outcomes with First-Line Nivolumab Plus Ipilimumab–Based Treatment in Patients with Metastatic NSCLC and Tumor Programmed Death-Ligand 1 Lower Than 1%: A Pooled Analysis. J. Thorac. Oncol. 2025, 20, 94–108. [Google Scholar] [CrossRef]
  81. Gray, J.E.; Villegas, A.; Daniel, D.; Vicente, D.; Murakami, S.; Hui, R.; Kurata, T.; Chiappori, A.; Lee, K.H.; Cho, B.C.; et al. Three-Year Overall Survival with Durvalumab after Chemoradiotherapy in Stage III NSCLC—Update from PACIFIC. J. Thorac. Oncol. 2020, 15, 288–293. [Google Scholar] [CrossRef] [PubMed]
  82. Barlesi, F.; Vansteenkiste, J.; Spigel, D.; Ishii, H.; Garassino, M.; de Marinis, F.; Özgüroğlu, M.; Szczesna, A.; Polychronis, A.; Uslu, R.; et al. Avelumab versus Docetaxel in Patients with Platinum-Treated Advanced Non-Small-Cell Lung Cancer (JAVELIN Lung 200): An Open-Label, Randomised, Phase 3 Study. Lancet Oncol. 2018, 19, 1468–1479, Erratum in Lancet Oncol. 2018, 19, e581. [Google Scholar] [CrossRef]
  83. Fares, C.M.; Van Allen, E.M.; Drake, C.G.; Allison, J.P.; Hu-Lieskovan, S. Mechanisms of Resistance to Immune Checkpoint Blockade: Why Does Checkpoint Inhibitor Immunotherapy Not Work for All Patients? Am. Soc. Clin. Oncol. Educ. B. 2019, 39, 147–164. [Google Scholar] [CrossRef]
  84. Nuti, M.; Zizzari, I.G.; Botticelli, A.; Rughetti, A.; Marchetti, P. The Ambitious Role of Anti Angiogenesis Molecules: Turning a Cold Tumor into a Hot One. Cancer Treat. Rev. 2018, 70, 41–46. [Google Scholar] [CrossRef]
  85. Socinski, M.A.; Jotte, R.M.; Cappuzzo, F.; Orlandi, F.; Stroyakovskiy, D.; Nogami, N.; Rodríguez-Abreu, D.; Moro-Sibilot, D.; Thomas, C.A.; Barlesi, F.; et al. Atezolizumab for First-Line Treatment of Metastatic Nonsquamous NSCLC. N. Engl. J. Med. 2018, 378, 2288–2301. [Google Scholar] [CrossRef]
  86. Bahnassy, A.A.; Ismail, H.; Mohanad, M.; El-Bastawisy, A.; Yousef, H.F. The Prognostic Role of PD-1, PD-L1, ALK, and ROS1 Proteins Expression in Non-Small Cell Lung Carcinoma Patients from Egypt. J. Egypt. Natl. Canc. Inst. 2022, 34, 23. [Google Scholar] [CrossRef]
  87. Rihawi, K.; Gelsomino, F.; Sperandi, F.; Melotti, B.; Fiorentino, M.; Casolari, L.; Ardizzoni, A. Pembrolizumab in the Treatment of Metastatic Non-Small Cell Lung Cancer: A Review of Current Evidence. Ther. Adv. Respir. Dis. 2017, 11, 353–373. [Google Scholar] [CrossRef]
  88. Li, X.; Wang, W.; Xu, C.; Yuan, Q. Efficacy and Safety of Immunotherapy Combined with Anlotinib as FirstLine Treatment in Older NSCLC Patients with PD-L1 Expression < 50%. Curr. Cancer Drug Targets 2025, 25, 818–828. [Google Scholar] [CrossRef] [PubMed]
  89. Pandey, R.; Shukla, S.; Husain, N.; Islam, M.; Hadi, R.; Kant Tripathi, S.; Singhal, A. Correlation Between Programmed Death Ligand-1(PD-L1) Expression and Driver Gene Mutations in Non-Small Cell Lung Carcinoma- Adenocarcinoma Phenotype. Asian Pac. J. Cancer Prev. 2022, 23, 131–142. [Google Scholar] [CrossRef] [PubMed]
  90. Gang, X.; Yan, J.; Li, X.; Shi, S.; Xu, L.; Liu, R.; Cai, L.; Li, H.; Zhao, M. Immune Checkpoint Inhibitors Rechallenge in Non-Small Cell Lung Cancer: Current Evidence and Future Directions. Cancer Lett. 2024, 604, 217241. [Google Scholar] [CrossRef] [PubMed]
  91. Sun, Q.; Wei, X.; Wang, Z.; Zhu, Y.; Zhao, W.; Dong, Y. Primary and Acquired Resistance against Immune Check Inhibitors in Non-Small Cell Lung Cancer. Cancers 2022, 14, 3294. [Google Scholar] [CrossRef]
  92. Lee, Y.J.; Lee, J.B.; Ha, S.-J.; Kim, H.R. Clinical Perspectives to Overcome Acquired Resistance to Anti–Programmed Death-1 and Anti–Programmed Death Ligand-1 Therapy in Non-Small Cell Lung Cancer. Mol. Cells 2021, 44, 363–373. [Google Scholar] [CrossRef]
  93. Lombardo, S.; Bramanti, A.; Ciurleo, R.; Basile, M.; Pennisi, M.; Bella, R.; Mangano, K.; Bramanti, P.; Nicoletti, F.; Fagone, P. Profiling of Inhibitory Immune Checkpoints in Glioblastoma: Potential Pathogenetic Players. Oncol. Lett. 2020, 20, 332. [Google Scholar] [CrossRef]
  94. Leone, G.M.; Mangano, K.; Caponnetto, S.; Fagone, P.; Nicoletti, F. Identification of Poliovirus Receptor-like 3 Protein as a Prognostic Factor in Triple-Negative Breast Cancer. Cells 2024, 13, 1299. [Google Scholar] [CrossRef]
  95. Patwekar, M.; Sehar, N.; Patwekar, F.; Medikeri, A.; Ali, S.; Aldossri, R.M.; Rehman, M.U. Novel Immune Checkpoint Targets: A Promising Therapy for Cancer Treatments. Int. Immunopharmacol. 2024, 126, 111186. [Google Scholar] [CrossRef] [PubMed]
  96. Borgeaud, M.; Sandoval, J.; Obeid, M.; Banna, G.; Michielin, O.; Addeo, A.; Friedlaender, A. Novel Targets for Immune-Checkpoint Inhibition in Cancer. Cancer Treat. Rev. 2023, 120, 102614. [Google Scholar] [CrossRef]
  97. Huang, Q.; Li, Y.; Huang, Y.; Wu, J.; Bao, W.; Xue, C.; Li, X.; Dong, S.; Dong, Z.; Hu, S. Advances in Molecular Pathology and Therapy of Non-Small Cell Lung Cancer. Signal Transduct. Target. Ther. 2025, 10, 186. [Google Scholar] [CrossRef] [PubMed]
  98. Wang, M.; Du, Q.; Jin, J.; Wei, Y.; Lu, Y.; Li, Q. LAG3 and Its Emerging Role in Cancer Immunotherapy. Clin. Transl. Med. 2021, 11, e365. [Google Scholar] [CrossRef]
  99. Shi, A.-P.; Tang, X.-Y.; Xiong, Y.-L.; Zheng, K.-F.; Liu, Y.-J.; Shi, X.-G.; Lv, Y.; Jiang, T.; Ma, N.; Zhao, J.-B. Immune Checkpoint LAG3 and Its Ligand FGL1 in Cancer. Front. Immunol. 2022, 12, 785091. [Google Scholar] [CrossRef]
  100. Qi, Y.; Chen, L.; Liu, Q.; Kong, X.; Fang, Y.; Wang, J. Research Progress Concerning Dual Blockade of Lymphocyte-Activation Gene 3 and Programmed Death-1/Programmed Death-1 Ligand-1 Blockade in Cancer Immunotherapy: Preclinical and Clinical Evidence of This Potentially More Effective Immunotherapy Strategy. Front. Immunol. 2021, 11, 563258. [Google Scholar] [CrossRef]
  101. 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] [PubMed]
  102. Krebs, M.G.; Forster, M.; Majem, M.; Peguero, J.; Iams, W.; Clay, T.; Roxburgh, P.; Doger, B.; Bajaj, P.; Barba, A.; et al. Eftilagimod Alpha (a Soluble LAG-3 Protein) Combined with Pembrolizumab in Second-Line Metastatic NSCLC Refractory to Anti–Programmed Cell Death Protein 1/Programmed Death-Ligand 1-Based Therapy: Final Results from a Phase 2 Study. JTO Clin. Res. Rep. 2024, 5, 100725. [Google Scholar] [CrossRef]
  103. 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]
  104. Zeidan, A.M.; Komrokji, R.S.; Brunner, A.M. TIM-3 Pathway Dysregulation and Targeting in Cancer. Expert Rev. Anticancer Ther. 2021, 21, 523–534. [Google Scholar] [CrossRef]
  105. Tang, R.; Rangachari, M.; Kuchroo, V.K. Tim-3: A Co-Receptor with Diverse Roles in T Cell Exhaustion and Tolerance. Semin. Immunol. 2019, 42, 101302. [Google Scholar] [CrossRef] [PubMed]
  106. Sabatos, C.A.; Chakravarti, S.; Cha, E.; Schubart, A.; Sánchez-Fueyo, A.; Zheng, X.X.; Coyle, A.J.; Strom, T.B.; Freeman, G.J.; Kuchroo, V.K. Interaction of Tim-3 and Tim-3 Ligand Regulates T Helper Type 1 Responses and Induction of Peripheral Tolerance. Nat. Immunol. 2003, 4, 1102–1110. [Google Scholar] [CrossRef] [PubMed]
  107. Curigliano, G.; Gelderblom, H.; Mach, N.; Doi, T.; Tai, D.; Forde, P.M.; Sarantopoulos, J.; Bedard, P.L.; Lin, C.-C.; Hodi, F.S.; et al. Phase I/Ib Clinical Trial of Sabatolimab, an Anti–TIM-3 Antibody, Alone and in Combination with Spartalizumab, an Anti–PD-1 Antibody, in Advanced Solid Tumors. Clin. Cancer Res. 2021, 27, 3620–3629, Erratum in Clin Cancer Res. 2024, 30, 3957. [Google Scholar] [CrossRef] [PubMed]
  108. Lin, C.-C.; Curigliano, G.; Santoro, A.; Kim, D.-W.; Tai, D.; Hodi, F.S.; Wilgenhof, S.; Doi, T.; Sabatos-Peyton, C.; Szpakowski, S.; et al. Sabatolimab in Combination with Spartalizumab in Patients with Non-Small Cell Lung Cancer or Melanoma Who Received Prior Treatment with Anti-PD-1/PD-L1 Therapy: A Phase 2 Multicentre Study. BMJ Open 2024, 14, e079132. [Google Scholar] [CrossRef]
  109. 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]
  110. Stanietsky, N.; Rovis, T.L.; Glasner, A.; Seidel, E.; Tsukerman, P.; Yamin, R.; Enk, J.; Jonjic, S.; Mandelboim, O. Mouse TIGIT Inhibits NK-cell Cytotoxicity upon Interaction with PVR. Eur. J. Immunol. 2013, 43, 2138–2150. [Google Scholar] [CrossRef]
  111. Yeo, J.; Ko, M.; Lee, D.-H.; Park, Y.; Jin, H. TIGIT/CD226 Axis Regulates Anti-Tumor Immunity. Pharmaceuticals 2021, 14, 200. [Google Scholar] [CrossRef]
  112. Qian, L.; Wu, L.; Miao, X.; Xu, J.; Zhou, Y. The Role of TIGIT-CD226-PVR Axis in Mediating T Cell Exhaustion and Apoptosis in NSCLC. Apoptosis 2025, 30, 784–804. [Google Scholar] [CrossRef]
  113. Chu, X.; Tian, W.; Wang, Z.; Zhang, J.; Zhou, R. Co-Inhibition of TIGIT and PD-1/PD-L1 in Cancer Immunotherapy: Mechanisms and Clinical Trials. Mol. Cancer 2023, 22, 93, Erratum in Mol. Cancer 2023, 22, 101. [Google Scholar] [CrossRef]
  114. Banta, K.L.; Xu, X.; Chitre, A.S.; Au-Yeung, A.; Takahashi, C.; O’Gorman, W.E.; Wu, T.D.; Mittman, S.; Cubas, R.; Comps-Agrar, L.; et al. Mechanistic Convergence of the TIGIT and PD-1 Inhibitory Pathways Necessitates Co-Blockade to Optimize Anti-Tumor CD8+ T Cell Responses. Immunity 2022, 55, 512–526.e9. [Google Scholar] [CrossRef] [PubMed]
  115. Zhang, P.; Liu, X.; Gu, Z.; Jiang, Z.; Zhao, S.; Song, Y.; Yu, J. Targeting TIGIT for Cancer Immunotherapy: Recent Advances and Future Directions. Biomark. Res. 2024, 12, 7. [Google Scholar] [CrossRef]
  116. 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]
  117. Khalaji, A.; Yancheshmeh, F.B.; Farham, F.; Khorram, A.; Sheshbolouki, S.; Zokaei, M.; Vatankhah, F.; Soleymani-Goloujeh, M. Don’t Eat Me/Eat Me Signals as a Novel Strategy in Cancer Immunotherapy. Heliyon 2023, 9, e20507. [Google Scholar] [CrossRef] [PubMed]
  118. Chen, Q.; Guo, X.; Ma, W. Opportunities and Challenges of CD47-Targeted Therapy in Cancer Immunotherapy. Oncol. Res. 2024, 32, 49–60. [Google Scholar] [CrossRef] [PubMed]
  119. Subbiah, V.; Vaishampayan, U.N.; Puri, S.; Kummar, S.; Perez, J.M.; Italiano, A.; Shao, J.; Bathia, R.; Sills, A.; Patel, S.P. A Phase 2 Multi-Arm Study of Magrolimab in Combination with Docetaxel in Patients with Locally Advanced or Metastatic Solid Tumors: ELEVATE Lung and UC. J. Clin. Oncol. 2023, 41, TPS9142. [Google Scholar] [CrossRef]
  120. 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]
  121. Garcia-Lorenzo, E.; Moreno, V. Targeting the Adenosine 2A Receptor in Non-Small Cell Lung Cancer: Shooting with Blank Bullets? Transl. Lung Cancer Res. 2023, 12, 653–656. [Google Scholar] [CrossRef]
  122. Van Kerkhove, O.; Verfaillie, S.; Maes, B.; Cuppens, K. The Adenosinergic Pathway in Non-Small Cell Lung Cancer. Cancers 2024, 16, 3142. [Google Scholar] [CrossRef] [PubMed]
  123. Sung, M.; Zer, A.; Walia, P.; Khoja, L.; Maganti, M.; Labbe, C.; Shepherd, F.A.; Bradbury, P.A.; Liu, G.; Leighl, N.B. Correlation of Immune-Related Adverse Events and Response from Immune Checkpoint Inhibitors in Patients with Advanced Non-Small Cell Lung Cancer. J. Thorac. Dis. 2020, 12, 2706–2712. [Google Scholar] [CrossRef]
  124. Blum, S.M.; Rouhani, S.J.; Sullivan, R.J. Effects of Immune-related Adverse Events (irAEs) and Their Treatment on Antitumor Immune Responses. Immunol. Rev. 2023, 318, 167–178. [Google Scholar] [CrossRef]
  125. Daniello, L.; Elshiaty, M.; Bozorgmehr, F.; Kuon, J.; Kazdal, D.; Schindler, H.; Shah, R.; Volckmar, A.-L.; Lusky, F.; Diekmann, L.; et al. Therapeutic and Prognostic Implications of Immune-Related Adverse Events in Advanced Non-Small-Cell Lung Cancer. Front. Oncol. 2021, 11, 703893. [Google Scholar] [CrossRef]
  126. Vaddepally, R.; Doddamani, R.; Sodavarapu, S.; Madam, N.R.; Katkar, R.; Kutadi, A.P.; Mathew, N.; Garje, R.; Chandra, A.B. Review of Immune-Related Adverse Events (IrAEs) in Non-Small-Cell Lung Cancer (NSCLC)—Their Incidence, Management, Multiorgan IrAEs, and Rechallenge. Biomedicines 2022, 10, 790. [Google Scholar] [CrossRef]
  127. Cufer, T.; Kosty, M.P. ESMO/ASCO Recommendations for a Global Curriculum in Medical Oncology Edition 2023. JCO Glob. Oncol. 2023, 9, e2300277. [Google Scholar] [CrossRef]
  128. Schneider, B.J.; Naidoo, J.; Santomasso, B.D.; Lacchetti, C.; Adkins, S.; Anadkat, M.; Atkins, M.B.; Brassil, K.J.; Caterino, J.M.; Chau, I.; et al. Management of Immune-Related Adverse Events in Patients Treated with Immune Checkpoint Inhibitor Therapy: ASCO Guideline Update. J. Clin. Oncol. 2021, 39, 4073–4126. [Google Scholar] [CrossRef] [PubMed]
  129. Gault, A.; Hogarth, L.; Williams, K.C.; Greystoke, A.; Rajan, N.; Speight, A.; Lamb, C.A.; Bridgewood, A.; Brown-Schofield, L.-J.; Rayner, F.; et al. Monitoring ImmunE DysregulAtion FoLLowing Immune CheckpOint-InhibitioN (MEDALLION): Protocol for an Observational Cancer Immunotherapy Cohort Study. BMC Cancer 2024, 24, 733. [Google Scholar] [CrossRef]
  130. Teimouri, A.; Minard, L.V.; Scott, S.N.; Daniels, A.; Snow, S. Real-World Adherence to Toxicity Management Guidelines for Immune-Related Adverse Events. Curr. Oncol. 2022, 29, 3104–3117. [Google Scholar] [CrossRef]
  131. Li, M.S.C.; Chan, A.L.S.; Mok, K.K.S.; Chan, L.L.; Mok, T.S.K. Next-Generation Immunotherapy: Igniting New Hope for Lung Cancer. Ther. Adv. Med. Oncol. 2024, 16, 17588359241302021. [Google Scholar] [CrossRef] [PubMed]
  132. Garg, P.; Pareek, S.; Kulkarni, P.; Horne, D.; Salgia, R.; Singhal, S.S. Next-Generation Immunotherapy: Advancing Clinical Applications in Cancer Treatment. J. Clin. Med. 2024, 13, 6537. [Google Scholar] [CrossRef] [PubMed]
  133. Punekar, S.R.; Shum, E.; Grello, C.M.; Lau, S.C.; Velcheti, V. Immunotherapy in Non-Small Cell Lung Cancer: Past, Present, and Future Directions. Front. Oncol. 2022, 12, 877594. [Google Scholar] [CrossRef]
  134. Kciuk, M.; Yahya, E.B.; Mohamed, M.M.I.; Rashid, S.; Iqbal, M.O.; Kontek, R.; Abdulsamad, M.A.; Allaq, A.A. Recent Advances in Molecular Mechanisms of Cancer Immunotherapy. Cancers 2023, 15, 2721. [Google Scholar] [CrossRef]
  135. Fu, Q.; Chen, N.; Ge, C.; Li, R.; Li, Z.; Zeng, B.; Li, C.; Wang, Y.; Xue, Y.; Song, X.; et al. Prognostic Value of Tumor-Infiltrating Lymphocytes in Melanoma: A Systematic Review and Meta-Analysis. Oncoimmunology 2019, 8, e1593806. [Google Scholar] [CrossRef] [PubMed]
  136. Vielva, L.R. Tumor Lymphocytic Infiltration in Non-Small Cell Lung Cancer: The Ultimate Prognostic Marker? Transl. Lung Cancer Res. 2016, 5, 370–372. [Google Scholar] [CrossRef] [PubMed]
  137. Liu, X.; Kong, Y.; Qian, Y.; Guo, H.; Zhao, L.; Wang, H.; Xu, K.; Ye, L.; Liu, Y.; Lu, H.; et al. Spatial Heterogeneity of Infiltrating Immune Cells in the Tumor Microenvironment of Non-Small Cell Lung Cancer. Transl. Oncol. 2024, 50, 102143. [Google Scholar] [CrossRef]
  138. Ellis, G.I.; Sheppard, N.C.; Riley, J.L. Genetic Engineering of T Cells for Immunotherapy. Nat. Rev. Genet. 2021, 22, 427–447. [Google Scholar] [CrossRef]
  139. Nix, M.A.; Wiita, A.P. Alternative Target Recognition Elements for Chimeric Antigen Receptor (CAR) T Cells: Beyond Standard Antibody Fragments. Cytotherapy 2024, 26, 729–738. [Google Scholar] [CrossRef]
  140. Roselli, E.; Boucher, J.C.; Li, G.; Kotani, H.; Spitler, K.; Reid, K.; Cervantes, E.V.; Bulliard, Y.; Tu, N.; Lee, S.B.; et al. 4-1BB and Optimized CD28 Co-Stimulation Enhances Function of Human Mono-Specific and Bi-Specific Third-Generation CAR T Cells. J. Immunother. Cancer 2021, 9, e003354. [Google Scholar] [CrossRef]
  141. Porter, D.L.; Levine, B.L.; Kalos, M.; Bagg, A.; June, C.H. Chimeric Antigen Receptor–Modified T Cells in Chronic Lymphoid Leukemia. N. Engl. J. Med. 2011, 365, 725–733. [Google Scholar] [CrossRef]
  142. Kalos, M.; Levine, B.L.; Porter, D.L.; Katz, S.; Grupp, S.A.; Bagg, A.; June, C.H. T Cells with Chimeric Antigen Receptors Have Potent Antitumor Effects and Can Establish Memory in Patients with Advanced Leukemia. Sci. Transl. Med. 2011, 3, 95ra73. [Google Scholar] [CrossRef] [PubMed]
  143. Sorkhabi, A.D.; Khosroshahi, L.M.; Sarkesh, A.; Mardi, A.; Aghebati-Maleki, A.; Aghebati-Maleki, L.; Baradaran, B. The Current Landscape of CAR T-Cell Therapy for Solid Tumors: Mechanisms, Research Progress, Challenges, and Counterstrategies. Front. Immunol. 2023, 14, 1113882. [Google Scholar] [CrossRef]
  144. Feng, K.; Guo, Y.; Dai, H.; Wang, Y.; Li, X.; Jia, H.; Han, W. Chimeric Antigen Receptor-Modified T Cells for the Immunotherapy of Patients with EGFR-Expressing Advanced Relapsed/Refractory Non-Small Cell Lung Cancer. Sci. China Life Sci. 2016, 59, 468–479. [Google Scholar] [CrossRef]
  145. Zhang, Y.; Zhang, Z.; Ding, Y.; Fang, Y.; Wang, P.; Chu, W.; Jin, Z.; Yang, X.; Wang, J.; Lou, J.; et al. Phase I Clinical Trial of EGFR-Specific CAR-T Cells Generated by the PiggyBac Transposon System in Advanced Relapsed/Refractory Non-Small Cell Lung Cancer Patients. J. Cancer Res. Clin. Oncol. 2021, 147, 3725–3734. [Google Scholar] [CrossRef]
  146. Fabrizio, F.P.; Muscarella, L.A.; Rossi, A. B7-H3/CD276 and Small-Cell Lung Cancer: What’s New? Transl. Oncol. 2024, 39, 101801. [Google Scholar] [CrossRef]
  147. Oh, M.S.; Abascal, J.; Rennels, A.K.; Salehi-Rad, R.; Dubinett, S.M.; Liu, B. Tumor Heterogeneity and the Immune Response in Non-Small Cell Lung Cancer: Emerging Insights and Implications for Immunotherapy. Cancers 2025, 17, 1027. [Google Scholar] [CrossRef]
  148. Andreou, T.; Neophytou, C.; Kalli, M.; Mpekris, F.; Stylianopoulos, T. Breaking Barriers: Enhancing CAR-Armored T Cell Therapy for Solid Tumors through Microenvironment Remodeling. Front. Immunol. 2025, 16, 1638186. [Google Scholar] [CrossRef]
  149. Xiao, X.; Huang, S.; Chen, S.; Wang, Y.; Sun, Q.; Xu, X.; Li, Y. Mechanisms of Cytokine Release Syndrome and Neurotoxicity of CAR T-Cell Therapy and Associated Prevention and Management Strategies. J. Exp. Clin. Cancer Res. 2021, 40, 367. [Google Scholar] [CrossRef] [PubMed]
  150. Martín-Rojas, R.M.; Gómez-Centurión, I.; Bailén, R.; Bastos, M.; Diaz-Crespo, F.; Carbonell, D.; Correa-Rocha, R.; Pion, M.; Muñoz, C.; Sancho, M.; et al. Hemophagocytic Lymphohistiocytosis/Macrophage Activation Syndrome (HLH/MAS) Following Treatment with Tisagenlecleucel. Clin. Case Rep. 2022, 10, e05209. [Google Scholar] [CrossRef] [PubMed]
  151. Sterner, R.C.; Sterner, R.M. CAR-T Cell Therapy: Current Limitations and Potential Strategies. Blood Cancer J. 2021, 11, 69. [Google Scholar] [CrossRef]
  152. Andersen, M.H. Tumor Microenvironment Antigens. Semin. Immunopathol. 2023, 45, 253–264. [Google Scholar] [CrossRef]
  153. Luo, J.; Zhang, X. Challenges and Innovations in CAR-T Cell Therapy: A Comprehensive Analysis. Front. Oncol. 2024, 14, 1399544. [Google Scholar] [CrossRef]
  154. Martín-Antonio, B.; Blanco, B.; González-Murillo, Á.; Hidalgo, L.; Minguillón, J.; Pérez-Chacón, G. Newer Generations of Multi-Target CAR and STAb-T Immunotherapeutics: NEXT CART Consortium as a Cooperative Effort to Overcome Current Limitations. Front. Immunol. 2024, 15, 1386856. [Google Scholar] [CrossRef] [PubMed]
  155. Zeltsman, M.; Dozier, J.; McGee, E.; Ngai, D.; Adusumilli, P.S. CAR T-Cell Therapy for Lung Cancer and Malignant Pleural Mesothelioma. Transl. Res. 2017, 187, 1–10. [Google Scholar] [CrossRef] [PubMed]
  156. Saffern, M.; Samstein, R. Taking CAR T Cells up a Synthetic Notch. Nat. Rev. Immunol. 2021, 21, 135. [Google Scholar] [CrossRef] [PubMed]
  157. Dimitri, A.; Herbst, F.; Fraietta, J.A. Engineering the Next-Generation of CAR T-Cells with CRISPR-Cas9 Gene Editing. Mol. Cancer 2022, 21, 78. [Google Scholar] [CrossRef]
  158. Wang, J.; Ma, J.; Xie, F.; Miao, F.; Lv, L.; Huang, Y.; Zhang, X.; Yu, J.; Tai, Z.; Zhu, Q.; et al. Immunogenic Cell Death-Based Cancer Vaccines: Promising Prospect in Cancer Therapy. Front. Immunol. 2024, 15, 1389173. [Google Scholar] [CrossRef]
  159. Liu, D.; Che, X.; Wang, X.; Ma, C.; Wu, G. Tumor Vaccines: Unleashing the Power of the Immune System to Fight Cancer. Pharmaceuticals 2023, 16, 1384. [Google Scholar] [CrossRef]
  160. Clinical Development and Perspectives of CIMAvax EGF, Cuban Vaccine for Non-Small-Cell Lung Cancer Therapy. MEDICC Rev. 2010, 12, 17. [CrossRef]
  161. Tagliamento, M.; Rijavec, E.; Barletta, G.; Biello, F.; Rossi, G.; Grossi, F.; Genova, C. CIMAvax-EGF, a Therapeutic Non-Small Cell Lung Cancer Vaccine. Expert Opin. Biol. Ther. 2018, 18, 829–835. [Google Scholar] [CrossRef]
  162. Vega, Y.I.F.; González, D.L.P.; Sarmiento, S.C.A.; Tul, L.E.A.; Valdés, I.B.I.; Machado, J.R.; Larrinaga, Á.E.; Rodríguez, J.E.F.; García, J.L.; Otero, D.C.; et al. Survival of NSCLC Patients Treated with Cimavax-EGF as Switch Maintenance in the Real-World Scenario. J. Cancer 2023, 14, 874–879. [Google Scholar] [CrossRef] [PubMed]
  163. Aurisicchio, L.; Ciliberto, G. Emerging Cancer Vaccines: The Promise of Genetic Vectors. Cancers 2011, 3, 3687–3713. [Google Scholar] [CrossRef] [PubMed]
  164. Tosch, C.; Bastien, B.; Barraud, L.; Grellier, B.; Nourtier, V.; Gantzer, M.; Limacher, J.M.; Quemeneur, E.; Bendjama, K.; Préville, X. Viral Based Vaccine TG4010 Induces Broadening of Specific Immune Response and Improves Outcome in Advanced NSCLC. J. Immunother. Cancer 2017, 5, 70. [Google Scholar] [CrossRef]
  165. Quoix, E.; Lena, H.; Losonczy, G.; Forget, F.; Chouaid, C.; Papai, Z.; Gervais, R.; Ottensmeier, C.; Szczesna, A.; Kazarnowicz, A.; et al. TG4010 Immunotherapy and First-Line Chemotherapy for Advanced Non-Small-Cell Lung Cancer (TIME): Results from the Phase 2b Part of a Randomised, Double-Blind, Placebo-Controlled, Phase 2b/3 Trial. Lancet Oncol. 2016, 17, 212–223. [Google Scholar] [CrossRef]
  166. Abakushina, E.V.; Popova, L.I.; Zamyatnin, A.A.; Werner, J.; Mikhailovsky, N.V.; Bazhin, A.V. The Advantages and Challenges of Anticancer Dendritic Cell Vaccines and NK Cells in Adoptive Cell Immunotherapy. Vaccines 2021, 9, 1363. [Google Scholar] [CrossRef]
  167. Madan, R.A.; Gulley, J.L. Sipuleucel-T: Harbinger of a New Age of Therapeutics for Prostate Cancer. Expert Rev. Vaccines 2011, 10, 141–150. [Google Scholar] [CrossRef]
  168. Lee, J.M.; Lee, M.-H.; Garon, E.; Goldman, J.W.; Salehi-Rad, R.; Baratelli, F.E.; Schaue, D.; Wang, G.; Rosen, F.; Yanagawa, J.; et al. Phase I Trial of Intratumoral Injection of CCL21 Gene–Modified Dendritic Cells in Lung Cancer Elicits Tumor-Specific Immune Responses and CD8+ T-Cell Infiltration. Clin. Cancer Res. 2017, 23, 4556–4568. [Google Scholar] [CrossRef]
  169. Salehi-Rad, R.; Lim, R.J.; Du, Y.; Tran, L.M.; Li, R.; Ong, S.L.; Ling Huang, Z.; Dumitras, C.; Zhang, T.; Park, S.J.; et al. CCL21-DC in Situ Vaccination in Murine NSCLC Overcomes Resistance to Immunotherapy and Generates Systemic Tumor-Specific Immunity. J. Immunother. Cancer 2023, 11, e006896. [Google Scholar] [CrossRef]
  170. Costin, D.; Chen, K.; Gralla, R.J.; Garofalo, B.; Wang, M.; Kurland, E. A Phase 2 Study of EIK1001, a Toll-like Receptor 7/8 (TLR7/8) Agonist, in Combination with Pembrolizumab and Chemotherapy in Patients with Stage 4 Non-Small Cell Lung Cancer. J. Clin. Oncol. 2024, 42, TPS8667. [Google Scholar] [CrossRef]
  171. Aggarwal, C.; Sterman, D.; Alesi, E.R.; Maldonado, F.; Mehra, R.; Bestvina, C.M.; Reisenauer, J.S.; Swartz, L.K.; Puri, S.; Ibrahim, O.; et al. Overall Survival after Treatment with CAN-2409 plus Valacyclovir in Combination with Continued ICI in Patients with Stage III/IV NSCLC with an Inadequate Response to ICI. J. Clin. Oncol. 2024, 42, 8634. [Google Scholar] [CrossRef]
  172. Hussain, M.S.; Sultana, A.; Bisht, A.S.; Gupta, G. Groundbreaking MRNA Lung Cancer Vaccine Trials: A New Dawn in Cancer Treatment. Curr. Cancer Drug Targets 2025, 25, 962–967. [Google Scholar] [CrossRef]
  173. Chen, D.-T.; Sabari, J.; Thompson, J.; Niu, J.; Mamdani, H.; Thapa, R.; Thompson, Z.; Posina, T.; Ryan, A.; Venhaus, R.; et al. Abstract CT052: A Phase IB Study of MRNA-Based Active Cancer Vaccine, BI1361849, Combined with Durvalumab and Tremelimumab Immunotherapy in Patients with Non-Small Cell Lung Cancer (NSCLC). Cancer Res. 2024, 84, CT052. [Google Scholar] [CrossRef]
  174. Lee, K.-W.; Yam, J.W.P.; Mao, X. Dendritic Cell Vaccines: A Shift from Conventional Approach to New Generations. Cells 2023, 12, 2147. [Google Scholar] [CrossRef]
  175. Yao, Y.; Fu, C.; Zhou, L.; Mi, Q.-S.; Jiang, A. DC-Derived Exosomes for Cancer Immunotherapy. Cancers 2021, 13, 3667. [Google Scholar] [CrossRef] [PubMed]
  176. Reynolds, G.; Tirard, A.; Villani, A.-C. Plasmacytoid Dendritic Cells: Welcome Back to the DC Fold. Immunity 2022, 55, 380–382. [Google Scholar] [CrossRef] [PubMed]
  177. Gu, Y.; Zhao, Q. Clinical Progresses and Challenges of Bispecific Antibodies for the Treatment of Solid Tumors. Mol. Diagn. Ther. 2024, 28, 669–702. [Google Scholar] [CrossRef]
  178. Herrera, M.; Pretelli, G.; Desai, J.; Garralda, E.; Siu, L.L.; Steiner, T.M.; Au, L. Bispecific Antibodies: Advancing Precision Oncology. Trends Cancer 2024, 10, 893–919. [Google Scholar] [CrossRef]
  179. Keam, S.J. Cadonilimab: First Approval. Drugs 2022, 82, 1333–1339. [Google Scholar] [CrossRef]
  180. Frentzas, S.; Gan, H.K.; Cosman, R.; Coward, J.; Tran, B.; Millward, M.; Zhou, Y.; Wang, W.; Xia, D.; Wang, Z.M.; et al. A Phase 1a/1b First-in-Human Study (COMPASSION-01) Evaluating Cadonilimab in Patients with Advanced Solid Tumors. Cell Rep. Med. 2023, 4, 101242. [Google Scholar] [CrossRef]
  181. Gao, X.; Xu, N.; Li, Z.; Shen, L.; Ji, K.; Zheng, Z.; Liu, D.; Lou, H.; Bai, L.; Liu, T.; et al. Safety and Antitumour Activity of Cadonilimab, an Anti-PD-1/CTLA-4 Bispecific Antibody, for Patients with Advanced Solid Tumours (COMPASSION-03): A Multicentre, Open-Label, Phase 1b/2 Trial. Lancet Oncol. 2023, 24, 1134–1146. [Google Scholar] [CrossRef]
  182. Huang, Y.; Yang, Y.; Zhao, Y.; Zhao, H.; Zhou, N.; Zhang, Y.; Chen, L.; Zhou, T.; Chen, G.; Wu, T.; et al. QL1706 (Anti-PD-1 IgG4/CTLA-4 Antibody) plus Chemotherapy with or without Bevacizumab in Advanced Non-Small Cell Lung Cancer: A Multi-Cohort, Phase II Study. Signal Transduct. Target. Ther. 2024, 9, 23. [Google Scholar] [CrossRef] [PubMed]
  183. Cho, B.C.; Simi, A.; Sabari, J.; Vijayaraghavan, S.; Moores, S.; Spira, A. Amivantamab, an Epidermal Growth Factor Receptor (EGFR) and Mesenchymal-Epithelial Transition Factor (MET) Bispecific Antibody, Designed to Enable Multiple Mechanisms of Action and Broad Clinical Applications. Clin. Lung Cancer 2023, 24, 89–97. [Google Scholar] [CrossRef]
  184. Liao, Y.-Y.; Tsai, C.-L.; Huang, H.-P. Optimizing Osimertinib for NSCLC: Targeting Resistance and Exploring Combination Therapeutics. Cancers 2025, 17, 459. [Google Scholar] [CrossRef]
  185. Zhou, C.; Tang, K.-J.; Cho, B.C.; Liu, B.; Paz-Ares, L.; Cheng, S.; Kitazono, S.; Thiagarajan, M.; Goldman, J.W.; Sabari, J.K.; et al. Amivantamab plus Chemotherapy in NSCLC with EGFR Exon 20 Insertions. N. Engl. J. Med. 2023, 389, 2039–2051. [Google Scholar] [CrossRef]
  186. Felip, E.; Cho, B.C.; Gutiérrez, V.; Alip, A.; Besse, B.; Lu, S.; Spira, A.I.; Girard, N.; Califano, R.; Gadgeel, S.M.; et al. Amivantamab plus Lazertinib versus Osimertinib in First-Line EGFR-Mutant Advanced Non-Small-Cell Lung Cancer with Biomarkers of High-Risk Disease: A Secondary Analysis from MARIPOSA. Ann. Oncol. 2024, 35, 805–816. [Google Scholar] [CrossRef] [PubMed]
  187. Rosenfeld, R. Amivantamab plus Lazertinib in Previously Untreated EGFR -Mutated Advanced NSCLC. N. Engl. J. Med. 2025, 392, 619–621. [Google Scholar] [CrossRef] [PubMed]
  188. Yu, H.A.; Chen, M.F.; Hui, A.B.; Choudhury, N.J.; Lee, J.J.-K.; Zheng, J.; Ahn, L.S.H.; Pupo, A.; Nesselbush, M.; Jabara, I.; et al. A Phase 2 Study of Amivantamab plus Lazertinib in Patients with EGFR- Mutant Lung Cancer and Active Central Nervous System Disease. J. Clin. Oncol. 2024, 42, 8517. [Google Scholar] [CrossRef]
  189. Cho, B.C.; Lu, S.; Felip, E.; Spira, A.I.; Girard, N.; Lee, J.-S.; Lee, S.-H.; Ostapenko, Y.; Danchaivijitr, P.; Liu, B.; et al. Amivantamab plus Lazertinib in Previously Untreated EGFR -Mutated Advanced NSCLC. N. Engl. J. Med. 2024, 391, 1486–1498. [Google Scholar] [CrossRef]
  190. Frentzas, S.; Austria Mislang, A.R.; Lemech, C.; Nagrial, A.; Underhill, C.; Wang, W.; Wang, Z.M.; Li, B.; Xia, Y.; Coward, J.I.G. Phase 1a Dose Escalation Study of Ivonescimab (AK112/SMT112), an Anti-PD-1/VEGF-A Bispecific Antibody, in Patients with Advanced Solid Tumors. J. Immunother. Cancer 2024, 12, e008037. [Google Scholar] [CrossRef]
  191. Zhao, Y.; Chen, G.; Chen, J.; Zhuang, L.; Du, Y.; Yu, Q.; Zhuang, W.; Zhao, Y.; Zhou, M.; Zhang, W.; et al. AK112, a Novel PD-1/VEGF Bispecific Antibody, in Combination with Chemotherapy in Patients with Advanced Non-Small Cell Lung Cancer (NSCLC): An Open-Label, Multicenter, Phase II Trial. eClinicalMedicine 2023, 62, 102106. [Google Scholar] [CrossRef]
  192. Dhillon, S. Ivonescimab: First Approval. Drugs 2024, 84, 1135–1142. [Google Scholar] [CrossRef] [PubMed]
  193. Duell, J.; Lammers, P.E.; Djuretic, I.; Chunyk, A.G.; Alekar, S.; Jacobs, I.; Gill, S. Bispecific Antibodies in the Treatment of Hematologic Malignancies. Clin. Pharmacol. Ther. 2019, 106, 781–791. [Google Scholar] [CrossRef] [PubMed]
  194. Salvador, J.-P.; Vilaplana, L.; Marco, M.-P. Nanobody: Outstanding Features for Diagnostic and Therapeutic Applications. Anal. Bioanal. Chem. 2019, 411, 1703–1713. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Timeline of the FDA approval of Immune Checkpoint Inhibitors (ICIs) for NSCLC treatment.
Figure 1. Timeline of the FDA approval of Immune Checkpoint Inhibitors (ICIs) for NSCLC treatment.
Ijms 26 11055 g001
Figure 2. Interaction of Immune Checkpoints and their corresponding ligands.
Figure 2. Interaction of Immune Checkpoints and their corresponding ligands.
Ijms 26 11055 g002
Figure 3. Autologous CAR-T cell manufacturing process. The production of autologous CAR T cells starts with the patient’s leukapheresis, followed by the enrichment and activation of T cells. The activated T lymphocytes are then transduced, for instance, using a lentiviral vector, to enable the introduction and, in some cases, permanent integration of the CAR transgene. The genetically modified T cells are subsequently expanded in either static or dynamic systems to reach the required therapeutic dose, cryopreserved, and reinfused into the patient.
Figure 3. Autologous CAR-T cell manufacturing process. The production of autologous CAR T cells starts with the patient’s leukapheresis, followed by the enrichment and activation of T cells. The activated T lymphocytes are then transduced, for instance, using a lentiviral vector, to enable the introduction and, in some cases, permanent integration of the CAR transgene. The genetically modified T cells are subsequently expanded in either static or dynamic systems to reach the required therapeutic dose, cryopreserved, and reinfused into the patient.
Ijms 26 11055 g003
Table 2. Ongoing clinical studies on anti-Lag3 therapies for NSCLC registered at clinicaltrials.gov (accessed on 10 November 2025).
Table 2. Ongoing clinical studies on anti-Lag3 therapies for NSCLC registered at clinicaltrials.gov (accessed on 10 November 2025).
NCT NumberInterventionsStudy StatusSponsorPhase
NCT03916627FianlimabActive not recruitingRegeneron PharmaceuticalsPhase 2
NCT05785767FianlimabRecruitingRegeneron PharmaceuticalsPhase 2|3
NCT05800015FianlimabRecruitingRegeneron PharmaceuticalsPhase 2|3
NCT06161441FianlimabRecruitingRegeneron PharmaceuticalsPhase 2
NCT06865339FianlimabRecruitingNitin OhriPhase 2
NCT06918132FianlimabRecruitingMayo ClinicPhase 2
NCT05978401GLS-012Not yet recruitingGuangzhou Gloria BiosciencesPhase 1|2
NCT05787613HLX26Active not recruitingShanghai Henlius BiotechPhase 2
NCT04205552RelatlimabRecruitingUniversity Hospital, EssenPhase 2
NCT04623775RelatlimabActive not recruitingBristol-Myers SquibbPhase 2
NCT05176483RelatlimabActive not recruitingExelixisPhase 1
NCT06561386RelatlimabRecruitingBristol-Myers SquibbPhase 3
Table 3. Ongoing clinical studies on anti-Tim3 therapies for NSCLC registered at clinicaltrials.gov (accessed on 10 November 2025).
Table 3. Ongoing clinical studies on anti-Tim3 therapies for NSCLC registered at clinicaltrials.gov (accessed on 10 November 2025).
NCT NumberInterventionsStudy StatusSponsorPhase
NCT02817633TSR-022Active not recruitingTesaro, Inc.Phase 1
NCT04931654AZD7789Active not recruitingAstraZenecaPhase 1|2
NCT06162572S095018RecruitingServier Bio-Innovation LLCPhase 1|2
Table 4. Ongoing clinical studies on anti-TIGIT therapies for NSCLC registered at clinicaltrials.gov (accessed on 10 November 2025).
Table 4. Ongoing clinical studies on anti-TIGIT therapies for NSCLC registered at clinicaltrials.gov (accessed on 10 November 2025).
NCT NumberStudy StatusInterventionsSponsorPhase
NCT03563716Active not recruitingTiragolumabGenentech, Inc.Phase 2
NCT04294810Active not recruitingTiragolumabHoffmann-La RochePhase 3
NCT04374877Active not recruitingCHS-388Coherus Biosciences, Inc.Phase 1
NCT04736173Active not recruitingDomvanalimabArcus Biosciences, Inc.Phase 2
NCT04746924Active not recruitingOciperlimabBeiGenePhase 3
NCT04995523RecruitingAZD2936AstraZenecaPhase 1|2
NCT05102214RecruitingHLX301Shanghai Henlius BiotechPhase 1|2
NCT05417321RecruitingHB0036Shanghai Huaota BiopharmaceuticalPhase 1|2
NCT05676931Active not recruitingDomvanalimabGilead SciencesPhase 2
NCT06627647RecruitingRilvegostomigAstraZenecaPhase 3
NCT06692738RecruitingRilvegostomigAstraZenecaPhase 3
NCT06773507RecruitingBC008-1ASichuan Luzhou Buchang BiopharmaceuticalPhase 1
Table 5. Ongoing clinical studies on CAR-T cell therapies for NSCLC registered at clinicaltrials.gov (accessed on 10 November 2025).
Table 5. Ongoing clinical studies on CAR-T cell therapies for NSCLC registered at clinicaltrials.gov (accessed on 10 November 2025).
NCT NumberStudy StatusInterventionsSponsorPhase
NCT04503278RecruitingCLDN6 CAR-TBioNTech Cell & Gene Therapies GmbHPhase 1
NCT04556669RecruitingaPD-L1 and anti-CD22 CAR-THebei Senlang Biotechnology Inc., Ltd.Phase 1
NCT05060796RecruitingCXCR5 modified EGFR CARTHospital of Guangzhou Medical UniversityPhase 1
NCT05239143Active not recruitingP-MUC1C-ALLO1 CAR-TPoseida Therapeutics, Inc.Phase 1
NCT05620342RecruitingiC9.GD2.CAR-TUNC Lineberger Comprehensive Cancer CenterPhase 1
NCT05736731Active not recruitingA2B530A2 Biotherapeutics Inc.Phase 1|2
NCT06043466RecruitingCEA-targeted CAR-TChongqing Precision Biotech Co., Ltd.Phase 1
NCT06051695RecruitingA2B694A2 Biotherapeutics Inc.Phase 1|2
NCT06653023RecruitingUniversal CAR-T injectionWondercel Biotech (ShenZhen)Phase 1
NCT06682793RecruitingA2B395A2 Biotherapeutics Inc.Phase 1|2
NCT06972576RecruitingEphA2-targeted CAR-T CellsZhejiang UniversityPhase 1
NCT07116057RecruitingMOv19-BBz CAR T cellsUniversity of PennsylvaniaPhase 1
Table 6. Ongoing clinical studies on oncological vaccines for NSCLC registered at clinicaltrials.gov (accessed on 10 November 2025).
Table 6. Ongoing clinical studies on oncological vaccines for NSCLC registered at clinicaltrials.gov (accessed on 10 November 2025).
NCT NumberStudy StatusInterventionsSponsorPhase
NCT01720836RecruitingMUC1 Peptide VaccineOlivera FinnPhase 1|2
NCT02432963Active not recruitingVirus Ankara Vaccine Expressing p53City of Hope Medical CenterPhase 1
NCT03546361Active not recruitingAutologous DC-Adenovirus CCL21 VaccineJonsson Comprehensive Cancer CenterPhase 1
NCT03552718Active not recruitingYE-NEO-001NantBioScience, Inc.Phase 1
NCT03908671RecruitingPersonalized mRNA Tumor VaccineStemirna TherapeuticsNot Applicable
NCT03970746Active not recruitingDC vaccinePDC*line Pharma SASPhase 1|2
NCT04147078RecruitingDC vaccineSichuan UniversityPhase 1
NCT04266730Not yet recruitingPANDA-VACUNC Lineberger Comprehensive Cancer CenterPhase 1
NCT04298606RecruitingEGF-rP64K/Montanide ISA 51 VaccineRoswell Park Cancer InstitutePhase 1
NCT04503278RecruitingCLDN6 uRNA-LPX/CLDN6 modRNA-LPXBioNTech Cell & Gene Therapies GmbHPhase 1
NCT04686305RecruitingT-DXdAstraZenecaPhase 1
NCT05104515RecruitingOVM-200Oxford Vacmedix UK Ltd.Phase 1
NCT05142189RecruitingBNT116BioNTech SEPhase 1
NCT05195619Active not recruitingPEP-DC vaccineCentre Hospitalier Universitaire VaudoisPhase 1
NCT05242965Active not recruitingPlasmid DNA VaccineUniversity of WashingtonPhase 2
NCT05254184RecruitingMutant KRAS-Targeted Peptide VaccineSidney Kimmel Comprehensive Cancer CenterPhase 1
NCT05269381RecruitingNeoantigen Peptide VaccineMayo ClinicPhase 1|2
NCT05344209RecruitingUV1Vestre Viken Hospital TrustPhase 2
NCT05557591Active not recruitingBNT116Regeneron PharmaceuticalsPhase 2
NCT05950139RecruitingPeptide vaccineSidney Kimmel Comprehensive Cancer CenterPhase 1|2
NCT06015724RecruitingKRAS vaccineGeorgetown UniversityPhase 2
NCT06095934RecruitingNeoantigen vaccineNanjing Drum Tower Hospital of Nanjing UniversityNot Applicable
NCT06253520RecruitingGRT-C903/GRT-R904National Cancer Institute (NCI)Phase 1
NCT06472245RecruitingOSE2101OSE ImmunotherapeuticsPhase 3
NCT06685653Not yet recruitingRGL-270Nanjing Tianyinshan HospitalPhase 1
NCT06735508Not yet recruitingmRNA Neoantigen VaccineGuangdong Provincial People’s HospitalPhase 1
NCT06751849RecruitingNeoantigen-loaded DC vaccineHospital of Nanchang UniversityPhase 2
NCT06751901RecruitingNeoantigen-based peptide vaccineHospital of Nanchang UniversityPhase 2
NCT06752044RecruitingNeoantigen-based peptide vaccineHospital of Nanchang UniversityNot Applicable
NCT06752057RecruitingNeoantigen-loaded DC vaccineHospital of Nanchang UniversityNot Applicable
NCT07073183Not yet recruitingCV09070101 mRNA vaccineCureVacPhase 1
Table 7. Ongoing clinical studies on bispecific antibody therapies for NSCLC registered at clinicaltrials.gov (accessed on 10 November 2025).
Table 7. Ongoing clinical studies on bispecific antibody therapies for NSCLC registered at clinicaltrials.gov (accessed on 10 November 2025).
NCT NumberStudy StatusInterventionsSponsorPhase
NCT02609776Active not recruitingAmivantamabJanssen Research & Development, LLCPhase 1
NCT02912949Active not recruitingMCLA-128Merus N.V.Phase 2
NCT03526835RecruitingMCLA-158Merus N.V.Phase 1|2
NCT03797391RecruitingEMB-01Shanghai EpimAb BiotherapeuticsPhase 1|2
NCT04140500Active not recruitingRO7247669Hoffmann-La RochePhase 1|2
NCT04603287Active not recruitingSI-B001Sichuan Baili PharmaceuticalPhase 1
NCT04606472Active not recruitingSI-B003Sichuan Baili PharmaceuticalPhase 1
NCT04777084RecruitingIBI318Hunan Province Tumor HospitalPhase 1
NCT04868877RecruitingMCLA-129Merus N.V.Phase 1|2
NCT04930432RecruitingMCLA-129Betta PharmaceuticalsPhase 1|2
NCT04931654Active not recruitingAZD7789AstraZenecaPhase 1|2
NCT04995523RecruitingAZD2936AstraZenecaPhase 1|2
NCT05102214RecruitingHLX301Shanghai Henlius BiotechPhase 1|2
NCT05117242Active not recruitingAcasunlimabGenmabPhase 2
NCT05180474Active not recruitingGEN1047GenmabPhase 1|2
NCT05360381Active not recruitingHLX35Shanghai Henlius BiotechPhase 1
NCT05377658RecruitingAK104Henan Cancer HospitalPhase 2
NCT05420220RecruitingKN046Jiangsu Alphamab BiopharmaceuticalsPhase 2
NCT05498389Not yet recruitingEMB-01Shanghai EpimAb BiotherapeuticsPhase 1|2
NCT05663866Active not recruitingAmivantamabJanssen Research & Development, LLCPhase 2
NCT05780307RecruitingIMM2520ImmuneOnco BiopharmaceuticalsPhase 1
NCT05816499Active not recruitingCadonilimabShanghai Chest HospitalPhase 1|2
NCT05845671RecruitingAmivantamabUniversity of Colorado, DenverPhase 1|2
NCT06015568Not yet recruitingMCLA-129Betta PharmaceuticalsPhase 1
NCT06116682RecruitingAmivantamabSWOG Cancer Research NetworkPhase 2
NCT06147037RecruitingFPI-2053Fusion Pharmaceuticals Inc.Phase 1
NCT06196814Not yet recruitingAK112Hunan Province Tumor HospitalPhase 1|2
NCT06361927RecruitingSSGJ-707Sunshine Guojian PharmaceuticalPhase 2
NCT06412471RecruitingSSGJ-707Sunshine Guojian PharmaceuticalPhase 2
NCT06417008RecruitingHS-20117Hansoh BioMedical R&D CompanyPhase 2|3
NCT06424821RecruitingCadonilimabShanghai Pulmonary HospitalPhase 2
NCT06467500RecruitingCadonilimabXin-Hua XuPhase 2
NCT06532591RecruitingCadonilimabSichuan Cancer Hospital and Research InstitutePhase 2
NCT06621563RecruitingHS-20117Hansoh BioMedical R&D CompanyPhase 1
NCT06724263Not yet recruitingB1962Tasly BiopharmaceuticalsPhase 2
NCT06766591Not yet recruitingIvonescimabJiangsu Province Nanjing Brain HospitalNot Applicable
NCT06793813RecruitingCadonilimabChinese Academy of Medical SciencesPhase 2
NCT06943820RecruitingAK129AkesoPhase 1|2
NCT06996782RecruitingRilvegostomigAstraZenecaPhase 1|2
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

Leone, G.M.; Scuderi, G.; Fagone, P.; Mangano, K. Directions of Immunotherapy for Non-Small-Cell Lung Cancer Treatment: Past, Present and Possible Future. Int. J. Mol. Sci. 2025, 26, 11055. https://doi.org/10.3390/ijms262211055

AMA Style

Leone GM, Scuderi G, Fagone P, Mangano K. Directions of Immunotherapy for Non-Small-Cell Lung Cancer Treatment: Past, Present and Possible Future. International Journal of Molecular Sciences. 2025; 26(22):11055. https://doi.org/10.3390/ijms262211055

Chicago/Turabian Style

Leone, Gian Marco, Grazia Scuderi, Paolo Fagone, and Katia Mangano. 2025. "Directions of Immunotherapy for Non-Small-Cell Lung Cancer Treatment: Past, Present and Possible Future" International Journal of Molecular Sciences 26, no. 22: 11055. https://doi.org/10.3390/ijms262211055

APA Style

Leone, G. M., Scuderi, G., Fagone, P., & Mangano, K. (2025). Directions of Immunotherapy for Non-Small-Cell Lung Cancer Treatment: Past, Present and Possible Future. International Journal of Molecular Sciences, 26(22), 11055. https://doi.org/10.3390/ijms262211055

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