Next Article in Journal
High-Efficiency Enrichment of Megakaryocytes and Identification of Micromegakaryocytes from Human Bone Marrow by Imaging Flow Cytometry
Previous Article in Journal
Hypothermia Shifts Neurodegeneration Phenotype in Neonatal Human Hypoxic–Ischemic Encephalopathy but Not in Related Piglet Models: Possible Relationship to Toxic Conformer and Intrinsically Disordered Prion-like Protein Accumulation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Resistance in Lung Cancer Immunotherapy and How to Overcome It: Insights from the Genetics Perspective and Combination Therapies Approach

1
Chair of Pathomorphology and Clinical Immunology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
2
Université Paris-Saclay, UVSQ, INSERM, END-ICAP, 94805 Versailles, France
3
Doctoral School, Medical University of Lublin, 20-954 Lublin, Poland
4
Faculty of Biology, University of Basel, 4123 Basel, Switzerland
5
Department of Endocrinology, Internal Medicine and Metabolism, Poznan University of Medical Sciences, 61-701 Poznan, Poland
6
Department of Pathomorphology, University Clinical Hospital, 61-701 Poznan, Poland
7
Department of Neurology, University Hospital Basel, 4123 Basel, Switzerland
8
Department of Biostatistics and Research Methodology, Faculty of Medicine, Collegium Medicum, Cardinal Stefan Wyszynski University of Warsaw, 02-004 Warsaw, Poland
9
Department of Histology and Embryology, Faculty of Medicine, Medical University of Warsaw, 02-004 Warsaw, Poland
10
Department of Methodology, Faculty of Medicine, Medical University of Warsaw, 02-004 Warsaw, Poland
11
IT Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland
*
Author to whom correspondence should be addressed.
Cells 2025, 14(8), 587; https://doi.org/10.3390/cells14080587
Submission received: 11 March 2025 / Revised: 7 April 2025 / Accepted: 9 April 2025 / Published: 12 April 2025
(This article belongs to the Special Issue Advances in Immunotherapy for Non-Small-Cell Lung Cancer)

Abstract

:
Lung cancer with the highest number of new cases diagnosed in Europe and in Poland, remains an example of malignancy with a very poor prognosis despite the recent progress in medicine. Different treatment strategies are now available for cancer therapy based on its type, molecular subtype and other factors including overall health, the stage of disease and cancer molecular profile. Immunotherapy is emerging as a potential addition to surgery, chemotherapy, radiotherapy or other targeted therapies, but also considered a mainstay therapy mode. This combination is an area of active investigation in order to enhance efficacy and overcome resistance. Due to the complexity and dynamic of cancer’s ecosystem, novel therapeutic targets and strategies need continued research into the cellular and molecular mechanisms within the tumour microenvironment. From the genetic point of view, several signatures ranging from a few mutated genes to hundreds of them have been identified and associated with therapy resistance and metastatic potential. ML techniques and AI can enhance the predictive potential of genetic signatures and model the prognosis. Here, we present the overview of already existing treatment approaches, the current findings of key aspects of immunotherapy, such as immune checkpoint inhibitors (ICIs), existing molecular biomarkers like PD-L1 expression, tumour mutation burden, immunoscore, and neoantigens, as well as their roles as predictive markers for treatment response and resistance.

1. Introduction

Lung cancer remains one of the most challenging malignancies worldwide, with particularly concerning statistics in Europe. Despite recent medical advances, mortality rates closely mirror incidence rates, with lung cancer deaths among women now exceeding breast cancer mortality [1]. In Europe, the annual incidence reaches 350,000 new cases in men (60,000 per 100,000) and 300,000 in women (50,000 per 100,000), with especially high rates in Eastern Europe, including Poland [2]. The percentage of different types of lung cancers is presented in Figure 1. While male incidence has begun to decline due to decreased smoking rates, the persistently high number of female smokers presents an ongoing concern, contributing to approximately 250,000 female and 300,000 male deaths annually in Europe [2].
In recent years, immunotherapy has revolutionised the treatment landscape for lung cancer, particularly through immune checkpoint inhibitors (ICIs). These agents work by stimulating the immune system to attack cancer cells, primarily through triggering and activating tumour-specific T cells [3]. The effectiveness of this approach relies on inducing or restoring the immune system’s capacity, especially T cells, to recognise and eliminate cancer cells [3]. This process involves a delicate balance known as cancer immunoediting, where the immune system both protects against tumour development and inadvertently promotes the survival of those cancer cells that escaped from immunosurveillance [3,4,5,6].
The success of immunotherapy, particularly PD-1/PD-L1 blockade, has been most pronounced in genetically complex tumours with high mutational loads, such as non-small cell lung cancer (NSCLC) [3,7]. Recent clinical trials have demonstrated significant advances, with the IMpower010 trial showing improved disease-free survival with adjuvant atezolizumab in resected NSCLC, and the CheckMate 816 trial establishing the benefits of neoadjuvant nivolumab combined with chemotherapy [8]. For small cell lung cancer (SCLC), the integration of ICIs into first-line chemotherapy regimens has become standard practice, with promising results from trials like ADRIATIC [9].
However, response rates to checkpoint inhibitors vary considerably, typically ranging from 8% to 30% [3,10,11,12,13]. While the durability of response is impressive—with 70–89% of responding patients maintaining remission for 6–14 months—these statistics highlight a critical challenge: many patients either fail to respond initially or develop resistance over time [3,10,11,12,13]. This variable response pattern occurs despite lung cancer’s relatively high mutation load, which theoretically provides numerous epitopes for cytotoxic T-cell targeting [14].
Furthermore, checkpoint inhibition can trigger a range of immune-related adverse events (irAEs) [13,15]. While most common side effects such as fatigue, skin rashes, and loss of appetite are manageable, more severe complications including autoimmune responses, pneumonitis, and even fatal pulmonary toxicity can occur in approximately 3% of anti-PD1-treated patients [15,16,17,18,19]. In extreme cases, immunotherapy may paradoxically induce hyperprogression of cancer, leading to rapid clinical deterioration [18,20,21,22].
Environmental factors, particularly tobacco smoking, significantly influence therapy success. Recent analyses have revealed correlations between smoking history and the expression of crucial immune-related genes, including the poliovirus receptor (PVR) and methylation patterns of PD-L1 and ICOSL genes [23,24,25]. These findings underscore the complex interplay between environmental factors and immune response in cancer treatment. To address these challenges, researchers are exploring various strategies, including novel checkpoint targets such as LAG-3, TIM-3, and OX-40 [26], combination therapies, and innovative delivery methods utilising nanomedicine [27]. Understanding and overcoming resistance mechanisms, particularly those associated with genetic mutations like STK11, KEAP1, and JAK1/2, has become crucial for improving patient outcomes [28].
In summary, the benefits of immunotherapy have changed the approach to treatment, especially in patients with NSCLC and SCLC. Patients who respond to treatment can experience remission for 6–14 months or longer [29]. Particularly promising results are observed in tumours with a high mutational burden, where immunotherapy can more effectively stimulate the immune system [30]. Groundbreaking studies such as Impower010 and Checkmate 816 have confirmed the efficacy of adjuvant and neoadjuvant therapy based on PD-1/PD-L1 inhibitors [31]. New strategies are being developed, including nanomedicine and combination therapies, that may increase the efficacy of immunotherapy [32]. Variable efficacy of immunotherapy—responses to ICIs range from 8% to 30%, meaning that many patients do not benefit from this therapy [33]. Some patients, despite an initial response, develop resistance to treatment, which limits the long-term efficacy of therapy. Smoking affects the expression of genes associated with the immune response (e.g., PVR, PD-L1, ICOSL), but the mechanisms of this impact require further study [34]. As an adverse event, immunotherapy can cause serious adverse events, such as autoimmune pneumonia or tumour hyperprogression, but the exact mechanisms of these complications are not yet fully understood. We need new therapeutic targets to identify alternative checkpoints (e.g., LAG-3, TIM-3, OX-40) and develop effective combination therapies [35].
This review focuses on the critical challenge of immunotherapy resistance in lung cancer, examining its mechanisms from a genetic perspective and exploring promising combination therapy approaches to overcome these limitations.

2. Treatment Landscape for Lung Cancer: Immunotherapy in Context

Treatment options for lung cancer include surgery, chemotherapy, radiotherapy, targeted therapies, immunotherapy, and various combinations of these approaches. In most lung cancer cases, immunotherapy has become the leading systemic treatment approach, second in importance only to surgery. This therapy works by leveraging our immune system’s fundamental ability to distinguish between the body’s own cells and foreign entities. Checkpoint inhibitor drugs, which counteract the mechanisms that cancer uses to evade immune detection, have shown exceptional promise in treatment. For this reason, our paper will concentrate primarily on immunotherapy as a lung cancer treatment strategy, while also describing how other therapeutic approaches can complement and enhance immunotherapy outcomes.

2.1. Immunotherapy

Immunotherapy, particularly checkpoint blockade therapies, has revolutionised lung cancer treatment by triggering and activating tumour-specific T cells [3]. This approach works through induction or restoration of the immune system’s capacity to recognise and eliminate cancer cells [3], operating within the framework of cancer immunoediting—a fragile balance where the immune system both protects against tumour development while inadvertently promoting the growth of those were able to escape from immunosurveillance [3,4,5,6]. Checkpoint inhibitors have shown particular efficacy in genetically complex tumours with high mutational loads and numerous neoantigens, such as NSCLC [3,7], with proven effectiveness in multiple cancer types including melanoma, head and neck cancer, and metastatic bladder cancer [10]. Despite their promise, response rates vary considerably from 8% to nearly 30%, though responding patients often maintain durable remission for 6–14 months [3,10,11,12,13]. This variable efficacy, despite lung cancer’s relatively high mutation burden [14], underscores our incomplete understanding of the immune synapse between tumour and immune cells. Immune-related adverse events remain a challenge, ranging from common side effects (fatigue, skin rashes) to serious complications like autoimmune responses and pneumonitis, with approximately 3% of anti-PD1-treated patients developing severe pulmonary toxicity [15,16,17,18,19]. Environmental factors, particularly tobacco smoking, significantly influence treatment outcomes by affecting the expression of immune-related genes such as PVR and methylation patterns of PD-L1 and ICOSL genes, highlighting the complex interplay between environmental exposures and immune response in cancer treatment [23,24,25].

2.2. Surgery

Surgery remains the primary intervention for early-stage disease (stages I-IIIa), particularly in NSCLC, offering the best chance for cure by physically removing cancerous tissue [36,37,38,39,40,41]. It eliminates immunosuppressive factors such as tumour-associated fibroblasts and extracellular matrix components while releasing tumour-specific antigens that trigger stronger T-cell responses [36,38]. The resulting inflammatory response mobilises immune cells to the area, creating favourable conditions for immune-mediated tumour control [38,39]. This post-surgical immune landscape offers a strategic opportunity for adjuvant immunotherapy to target microscopic residual disease with enhanced efficacy.

2.3. Chemotherapy

Chemotherapy, once the standard treatment for advanced lung cancer, now demonstrates its greatest value in combination with immunotherapy [42]. This synergistic approach potentiates immunotherapy effects by increasing tumour antigenicity and disrupting the immunosuppressive tumour microenvironment [43]. Pivotal studies have established the clinical benefit of these combinations. Gandhi et al. (2018) demonstrated that adding pembrolizumab to platinum-based chemotherapy with pemetrexed significantly improved overall survival, progression-free survival, and response rates in patients with metastatic non-squamous NSCLC without EGFR or ALK alterations [44]. Similarly, Socinski et al. (2018) showed that atezolizumab combined with bevacizumab and chemotherapy improved survival outcomes regardless of PD-L1 expression or genetic alteration status [45]. These landmark studies have transformed the first-line treatment paradigm for advanced NSCLC without targetable mutations, establishing chemoimmunotherapy combinations as the standard of care. The immunomodulatory effects of cytotoxic agents appear to enhance immune recognition of tumour cells while simultaneously reducing immunosuppressive elements within the tumour microenvironment, creating conditions more favourable for effective immune surveillance and attack.

2.4. Radiotherapy

Radiotherapy, one of the oldest cancer treatment modalities [46], has evolved from a primarily local tumour control strategy to a potential systemic immune modulator when combined with immunotherapy. While traditional radiation effects include direct DNA damage [47,48] and cell cycle disruption, its immunomodulatory effects have gained significant attention. Radiation enhances the recognition of tumour-associated antigens by the immune system, increasing the recruitment and activation of immune cells [49], which can prime the immune system for checkpoint inhibitor therapy. However, radiation’s immune effects are complex, as it can also induce immunosuppression through increased regulatory T cells and myeloid-derived suppressor cells [50].
Particularly relevant to immunotherapy integration is radiation’s ability to modulate PD-L1 expression, directly affecting tumour responsiveness to checkpoint inhibitors [51]. The combination of radiotherapy with immunotherapy has shown promising clinical results, as demonstrated by Theelen et al. (2021), who found improved overall survival in NSCLC patients receiving radiotherapy with pembrolizumab [52]. This approach can potentially induce the abscopal effect, where radiation-enhanced immune responses affect non-irradiated metastatic sites, transforming a local treatment into a systemic therapy. By strategically sequencing radiation with immunotherapy, clinicians can potentially leverage both the direct cytotoxic effects of radiation and its immune-stimulating properties to overcome resistance mechanisms and enhance treatment outcomes.

2.5. Targeted Therapies

Targeted therapies have transformed lung cancer treatment by precisely inhibiting specific oncogenic drivers like EGFR, ALK, ROS1, and KRAS G12C mutations, as indicated in Table 1 and Table 2 below [53,54]. Despite impressive initial responses in patients with actionable mutations, resistance inevitably develops, necessitating alternative strategies [55,56]. The integration of targeted therapies with immunotherapy represents an emerging approach to overcome resistance mechanisms and extend treatment benefits. Studies by Nakagawa et al. (2019) demonstrated that dual blockade of the EGFR and VEGF pathways using EGFR TKIs with ramucirumab (a VEGFR2 antagonist) improved progression-free survival in EGFR-mutated metastatic NSCLC [57].
This combination approach addresses a significant challenge: patients with oncogene-driven tumours typically show lower response rates to immune checkpoint inhibitors as monotherapy, likely due to lower tumour mutational burden and less immunogenic microenvironments [3,7]. By strategically combining targeted agents with immunotherapy, treatment can simultaneously address the oncogenic driver while enhancing immune recognition and attack [58,59]. Emerging evidence suggests that certain targeted therapies may favourably alter the tumour immune microenvironment, potentially sensitising previously resistant tumours to immunotherapy [56,60].
The extensive range of FDA and EMA-approved targeted therapies for specific genetic alterations [59], including newer agents for previously “undruggable” targets like KRAS G12C, provides multiple opportunities for rational combination strategies [55,61]. As research advances, optimising the sequencing, dosing, and selection of specific combinations based on molecular profiles will be crucial for maximising efficacy while managing overlapping toxicities. This precision medicine approach, integrating both targeted therapy and immunotherapy based on comprehensive genetic profiling, represents the frontier of personalised treatment for lung cancer patients [45,53,54,55,56,58,59,60,61,62].
Table 1. Targeted therapies approved by the FDA and European Medicines Agency (EMA), available in Poland versus selected EU countries, designated for lung cancer treatment—included in the national oncologic treatment schemes [59].
Table 1. Targeted therapies approved by the FDA and European Medicines Agency (EMA), available in Poland versus selected EU countries, designated for lung cancer treatment—included in the national oncologic treatment schemes [59].
Drug Name and Availability in Poland Versus in Selected EU CountriesPolandItalySpainSwedenSloveniaCzechiaEstonia
Osimertinib
2nd line post EGFR TKI, T790M+
Osimertinib
1st line, EGFR M+
Crizotinib
1st line, ALK+ patients
Crizotinib
any line, ROS+ patients
Alectinib
1st line, ALK+ patients
Dabrafenib-trametinib
BRAF+ patients
Green = widely available. Yellow = available with restriction. Red = not available.
Table 2. Selected molecularly targeted therapies approved by the FDA and/or EMA for use in lung cancer or any cancer as long as they possess certain genetic variants (so-called oncoagnostic approach, in line with modern precision medicine), often used in combination therapies. It is important to bear in mind that this table presents only some of the most prominent targeted therapies and mutations. The landscape continues to evolve with ongoing research and clinical trials [45,52,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112].
Table 2. Selected molecularly targeted therapies approved by the FDA and/or EMA for use in lung cancer or any cancer as long as they possess certain genetic variants (so-called oncoagnostic approach, in line with modern precision medicine), often used in combination therapies. It is important to bear in mind that this table presents only some of the most prominent targeted therapies and mutations. The landscape continues to evolve with ongoing research and clinical trials [45,52,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112].
DrugTargeted MutationFDA ApprovedAvailable in EUStage in Clinical Trials (If Not FDA Approved)
GefitinibEGFR exon 19 deletion/L858R mutationYesYes-
ErlotinibEGFR exon 19 deletion/L858R mutationYesYes-
OsimertinibEGFR T790M resistance mutationYesYes-
AlectinibALK rearrangementsYesYes-
BrigatinibALK rearrangementsYesYes-
CrizotinibALK, ROS1 rearrangementsYesYes-
LorlatinibALK rearrangementsYesYes-
SotorasibKRAS G12C mutationYesYes-
AdagrasibKRAS G12C mutationYesYes-
SelpercatinibRET fusionsYesYes-
PralsetinibRET fusionsYesYes-
CapmatinibMET exon 14 skipping mutationYesYes-
TepotinibMET exon 14 skipping mutationYesYes-
AmivantamabEGFR exon 20 insertion mutationYesYes-
MobocertinibEGFR exon 20 insertion mutationYesYes-
EntrectinibROS1, neurotrophic tyrosine receptor kinase (NTRK) gene fusionsYesYes-
LarotrectinibNTRK gene fusionsYesYes-
NivolumabPD-1 (immune checkpoint inhibitor)YesYes-
PembrolizumabPD-1 (immune checkpoint inhibitor)YesYes-
AtezolizumabPD-L1 (immune checkpoint inhibitor)YesYes-
DurvalumabPD-L1 (immune checkpoint inhibitor)YesYes-
SavolitinibMET exon 14 skipping mutationNoNoPhase 2/3
Dabrafenib/TrametinibBRAF V600E mutationYesYes-

2.6. Novel Immunotherapeutic Approaches

The evolution of immunotherapy extends beyond PD-1/PD-L1 and CTLA-4 inhibitors to novel checkpoint targets that address resistance mechanisms in lung cancer. Emerging approaches focus on alternative immune checkpoints including LAG-3, TIM-3, and TIGIT [113]. LAG-3 inhibitors like relatlimab combined with nivolumab have demonstrated promising activity in PD-L1-positive NSCLC patients, improving progression-free survival, while eftilagimod alpha shows potential in enhancing pembrolizumab efficacy by activating antigen-presenting cells [114,115]. TIM-3 targeting agents such as sabatolimab and TSR-022 have shown manageable toxicity profiles with improved outcomes when combined with PD-1 inhibitors, though with limited efficacy as monotherapies [26,116]. TIGIT blockade represents another promising pathway, with tiragolumab significantly enhancing progression-free survival when combined with atezolizumab in PD-L1-positive patients, while domvanalimab with zimberelimab has shown encouraging response rates [117,118]. Therapeutic cancer vaccines [119] and innovative combinations like BRAF and MEK inhibitors for BRAF V600E-mutant NSCLC [112] further expand treatment options. These novel approaches emphasise the importance of combinatorial strategies, as most new checkpoint inhibitors demonstrate modest activity alone but enhanced efficacy when integrated with established immunotherapies, targeted agents, or conventional treatments, providing promising avenues to overcome the complex resistance mechanisms in lung cancer [26,113,114,115,116,117,118,120]. Table 3 summarises the key findings from early-phase clinical trials investigating checkpoint inhibitors targeting LAG-3, TIM-3, TIGIT, and ICOS pathways.

3. Problem of Resistance to Immunotherapy in Lung Cancer

Immunotherapy, particularly immune checkpoint blockade, has revolutionised cancer treatment, including non-small cell lung cancer. However, resistance to immune checkpoint blockade remains a significant challenge, limiting its therapeutic potential for many patients [121]. Understanding these resistance patterns and their underlying mechanisms is crucial for developing effective treatment strategies.

3.1. Types of Resistance

Primary resistance refers to the lack of initial response to immunotherapy, occurring in 40–44% of NSCLC patients treated with immune checkpoint inhibitors as second-line therapy and 21–27% as first-line therapy [122]. The incidence is notably lower (around 10%) when ICIs are combined with chemotherapy as first-line treatment [44,123]. Primary resistance may result from various factors, including low tumour immunogenicity, impaired antigen presentation, and an immunosuppressive tumour microenvironment (TME) [124,125].
Secondary (acquired) resistance develops in patients who initially respond to immunotherapy but later experience disease progression [121]. This affects a significant number of NSCLC patients, with studies reporting secondary resistance rates of 52–57% for first-line and 32–64% for second-line ICI treatment [126]. The mechanisms underlying secondary resistance are complex and may involve changes in tumour cells, the TME, or both [127]. Key mechanisms of resistance include tumour-intrinsic factors such as loss of tumour antigens and neoantigen expression, alterations in antigen presentation machinery, activation of oncogenic signalling pathways, impaired interferon (IFN)-γ signalling, and upregulation of alternative immune checkpoints [128,129].

3.2. Mechanisms of Resistance

The mechanisms of resistance to immunotherapy in lung cancer are multifaceted, encompassing genetic alterations, metabolic changes, and environmental factors. Among these, specific genetic mutations play a particularly significant role in determining treatment outcomes.

3.2.1. Genomic Alterations and Associated Factors

Genetic alterations in lung cancer not only affect direct immune responses but also influence metabolic, epigenetic, and microbiome-mediated mechanisms of resistance.
STK11 (LKB1) Mutations
STK11 mutations represent a major challenge in immunotherapy resistance, associated with a non-T cell-inflamed TME and frequently co-occurring with KRAS mutations [130,131]. These mutations significantly impact cellular metabolism, leading to increased lactate production and secretion via the MCT4 transporter, which subsequently suppresses antitumour immunity by promoting M2 macrophage polarization and T-cell dysfunction [132]. The impact of STK11 mutations extends beyond direct metabolic effects, as they also influence epigenetic regulation. DNA methylation and histone modifications can further suppress the expression of genes involved in immune activation, exacerbating resistance to ICIs. This complex interaction suggests potential therapeutic opportunities through DNA methyltransferase inhibitors (DNMTis) or histone deacetylase inhibitors (HDACis) [132].
KEAP1 Mutations
KEAP1 mutations present another significant challenge, resulting in hyperactivation of the NRF2 pathway and leading to enhanced oxidative stress resistance and immune evasion [133]. These mutations profoundly affect cellular metabolism, increasing the consumption of glucose and glutamine by tumour cells, thereby depriving effector T cells of essential nutrients and promoting immunosuppression. Recent research has shown promising results in combining ICIs with glutaminase inhibitors to reverse these immunosuppressive effects [134,135,136]. The impact of KEAP1 mutations is further modulated by epigenetic regulation of the NRF2 pathway and its downstream targets, suggesting that targeting these epigenetic mechanisms might enhance immunotherapy efficacy [133].
JAK1/2 Mutations
JAK1/2 mutations disrupt interferon-gamma signalling, significantly impairing antigen presentation and T cell activation [130]. This disruption leads to metabolic dysregulation affecting cell surface marker expression and immune surveillance. Current research is exploring strategies to restore or bypass these pathways, such as using agents that enhance antigen presentation or T cell activation, to improve outcomes in JAK1/2-mutant tumours [137,138]. These mutations also influence epigenetic regulation, particularly in the downregulation of interferon-stimulated genes, contributing to resistance mechanisms [139].
The above-described mutations have been summarised in Table 4 below.

3.2.2. Microbiome Influence

The gut microbiome plays a crucial role in modulating responses to immunotherapy across all these genetic contexts. Research has shown that specific bacterial species and overall microbiome diversity can significantly impact ICI efficacy. In patients with STK11 mutations, the immunosuppressive TME can be influenced by the gut microbiome, suggesting potential therapeutic approaches through microbiome modulation [140]. Similarly, the efficacy of metabolic interventions in KEAP1-mutant tumours may be modulated by the gut microbiome [136], while microbiome-based interventions might help restore immune function in JAK1/2-mutant cases [141].

3.2.3. Tumour Microenvironment Factors

The tumour microenvironment (TME) of lung cancer represents a complex and dynamic ecosystem that significantly influences immunotherapy outcomes [142]. The complexity of TME is illustrated in Figure 2. Understanding its components and their interactions is crucial for comprehending resistance mechanisms and developing effective therapeutic strategies.
Cancer-Associated Fibroblasts (CAFs)
Cancer-associated fibroblasts serve as a major component of the TME, playing a significant role in promoting therapy resistance. These cells secrete extracellular matrix (ECM) components and remodelling enzymes that facilitate tumour progression and treatment resistance through ECM modification. Additionally, CAFs release critical signalling molecules, including TGF-β, VEGF, and IL-6, which actively modulate immune responses and angiogenesis [143]. The targeting of these stromal components has emerged as a promising strategy to combat tumour development and resistance to conventional therapies, potentially transforming lung cancer into a more manageable disease [144].
Immune Cell Composition
The immune landscape within lung cancer presents a diverse array of cells that influence treatment response. This includes tumour-associated macrophages, which differentiate into either proinflammatory M1 (anti-tumour) or immunosuppressive M2 (pro-tumour) phenotypes, T cells, natural killer (NK) cells, and myeloid-derived suppressor cells (MDSCs). The presence of regulatory T cells (Tregs) and exhausted CD8+ T cells within the TME frequently leads to compromised anti-tumour immunity [145]. A “T cell-inflamed” TME, characterised by high CD8+ T cell infiltration, is typically associated with better responses to immune checkpoint inhibitors, while an immunosuppressive environment dominated by regulatory T cells, MDSCs, and M2 macrophages often leads to treatment resistance [146].
Hypoxia and Angiogenesis
Hypoxia represents a critical feature of the TME that contributes to immunotherapy resistance. The stabilization of HIF-1α under hypoxic conditions regulates genes involved in angiogenesis, metabolism, and survival, promoting VEGF expression and correlating with poor prognosis in lung cancer patients [147]. Endothelial cells play a vital role in this process through the formation of new blood vessels, with VEGF serving as a key mediator. Beyond VEGF, other angiogenic factors including FGF-2 and HIFs contribute to tumour cell proliferation, migration, and invasion [148,149]. Strategies to alleviate hypoxia, such as HIF-1α inhibitors or agents that normalise tumour vasculature, may enhance the effectiveness of immunotherapy by improving immune cell infiltration and activity [150].
Extracellular Matrix and Signalling Pathways
The ECM provides both structural support and regulatory functions through biochemical and mechanical signals. In lung cancer, ECM dysregulation promotes tumour cell proliferation, migration, and invasion. Matrix metalloproteinases (MMPs) and other enzymes, including lysyl oxidases and ADAMTS, facilitate metastasis through ECM remodelling [151]. The TGF-β signalling pathway particularly influences immunotherapy resistance by inducing epithelial-mesenchymal transition, enhancing invasion, and suppressing immune responses, contributing to a more aggressive phenotype [152].

3.2.4. Comorbidities Impact

Comorbidities significantly influence the efficacy of immunotherapy, particularly in both NSCLC and SCLC patients. The introduction of immunotherapy, particularly immune checkpoint inhibitors (ICIs), has significantly transformed the therapeutic landscape of lung cancer treatment. ICIs, which primarily function by inhibiting the interaction between PD-1/PD-L1 or CTLA-4 and their respective ligands, have redefined oncological treatment protocols. While immunotherapy offers promising clinical benefits, it is also associated with immune-related adverse events (irAEs), a distinct spectrum of treatment-related complications. Among these, immune-related pneumonitis (IRP) is of particular concern, especially in patients with lung cancer. Although IRP remains a relatively rare occurrence, its potential severity—ranging from mild, manageable symptoms to life-threatening respiratory failure—necessitates careful clinical consideration [153].
Chronic Obstructive Pulmonary Disease (COPD) is a prevalent respiratory condition characterised by persistent symptoms and airflow limitation. It is a heterogeneous disorder encompassing various phenotypes, including emphysema and chronic bronchitis. The overlap between COPD and lung cancer is substantial, with shared risk factors such as smoking and environmental exposures contributing to chronic inflammation and cellular alterations that predispose individuals to oncogenesis. These complexities render COPD a critical factor in the management of lung cancer, particularly in relation to IRP development during ICI therapy. The pathogenesis of COPD involves chronic airway inflammation, structural remodelling, and significant alterations in pulmonary immune responses. These mechanisms may predispose COPD patients to an elevated risk of IRP when treated with ICIs, given their already compromised pulmonary function. A pooled analysis has demonstrated that lung cancer patients with concomitant COPD exhibit a heightened susceptibility to IRP during immunotherapy. Fangyuan presents compelling evidence indicating a significantly elevated risk of immune-related pneumonitis (IRP) in patients with concomitant Chronic Obstructive Pulmonary Disease (COPD). IRP is a potentially life-threatening complication that can severely impair respiratory function and diminish quality of life, often necessitating the discontinuation of life-prolonging immunotherapy. Among the identified risk factors for IRP, COPD has emerged as a critical contributor. Patients with COPD exhibit a heightened susceptibility to IRP due to pre-existing pulmonary inflammation and an already compromised immune response, which may interact with the immunomodulatory effects of immune checkpoint inhibitors (ICIs) [154]. Zhou’s findings underscore the impact of COPD on the risk of IRP in lung cancer patients undergoing immunotherapy. By identifying this high-risk subgroup, oncologists can more effectively weigh the risks and benefits of immunotherapy, facilitating the development of predictive models for IRP and the implementation of preventive strategies to mitigate its occurrence. The identification of high-risk subgroups among lung cancer patients is crucial for optimising the balance between the risks and benefits of immunotherapy. By recognising such populations, oncologists can facilitate the development of predictive models for immune-related pneumonitis (IRP) and implement preventive strategies to mitigate its occurrence.
Chronic obstructive pulmonary disease (COPD) presents a complex interplay with immunotherapy response. Patients with COPD exhibit increased Th1 differentiation in both pulmonary and tumour tissues, potentially influencing the efficacy of immune checkpoint inhibitors (ICIs) [155]. Additionally, the presence of multiple comorbidities, as assessed by the Charlson Comorbidity Index (CCI), has been linked to poorer progression-free survival in individuals undergoing PD-1 inhibitor therapy [156].
The significant heterogeneity observed in studies investigating this relationship may be attributable to variations in study populations, lung cancer types and stages, COPD severity, and differing immunotherapeutic regimens. The elevated odds ratio (OR) highlights a critical clinical consideration for practitioners managing lung cancer patients with pre-existing COPD, as these individuals face a heightened risk of developing IRP—a potentially severe and treatment-limiting adverse event. This increased susceptibility may stem from the underlying inflammatory state and altered pulmonary immune environment associated with COPD, which could interact with the mechanisms of action of ICIs, leading to an exacerbated inflammatory response in the lungs [154].
Furthermore, lung cancer patients with COPD are at an elevated risk of IRP, particularly in the context of radiation therapy. Notably, this heightened risk persists across varying levels of radiation exposure (< 40% and ≥ 40%), suggesting that COPD functions as an independent risk factor for IRP. When compounded by radiation-induced effects, the likelihood of this adverse event may be further increased.
The literature presents conflicting findings regarding the influence of COPD on lung cancer treatment outcomes. While some studies associate COPD with poorer responses to ICIs, others indicate that in patients with COPD, Th1 cell populations are expanded in both the lung and tumour microenvironments. Notably, COPD has been linked to prolonged progression-free intervals in patients treated with ICIs [155]. These findings underscore the need for further investigation into the immune mediators of COPD and their implications for developing novel therapeutic strategies for non-small cell lung cancer (NSCLC).
The impact of comorbidities is particularly noteworthy in small-cell lung cancer (SCLC) patients. While these individuals often exhibit a high mutational burden due to smoking history, the presence of comorbidities such as renal or hepatic impairment can significantly complicate treatment outcomes. The American Society of Clinical Oncology (ASCO) guidelines specifically highlight that patients with poor performance status or significant comorbidities tend to have inferior outcomes with dual ICI therapy, likely due to the poor prognosis associated with their underlying conditions [157]. This challenge is further compounded by the immunosuppressive phenotype of SCLC, characterised by low PD-L1 expression and poor T-cell infiltration, which adds another layer of complexity to ICI efficacy in these patients [158].

3.3. Patient Characteristics Associated with Resistance

Primary resistance patterns are particularly evident in specific patient populations. Non-smokers tend to show higher rates of primary resistance, primarily due to their typically lower tumour mutational burden, which correlates with reduced immunogenicity and poor response to ICIs [159]. Patients with more extensive disease burden also frequently exhibit primary resistance, as the extensive disease can create an immunosuppressive tumour microenvironment that hinders effective immune responses. Similarly, patients with low albumin levels, which may reflect poor overall health and a compromised immune system, often show reduced efficacy of immunotherapy [159]. A crucial factor in primary resistance is poor T-cell infiltration, as tumours lacking sufficient CD8+ T-cell infiltration are less likely to respond to ICIs due to insufficient immune activation [160].
Acquired resistance presents a different set of challenges, developing in patients who initially respond to ICIs but later experience disease progression. This form of resistance often involves complex genomic alterations, with mutations in genes such as STK11, B2M, APC, MTOR, KEAP1, and JAK1/2 being implicated in resistance mechanisms [130]. The tumour microenvironment undergoes significant changes in cases of acquired resistance, characterised by increased infiltration of immunosuppressive cells like myeloid-derived suppressor cells and M2 macrophages, along with decreased neoantigen presentation [160]. Additionally, immunophenotypic changes play a crucial role, with some patients showing reduced numbers of tumour-infiltrating CD8+ T cells and decreased PD-L1 expression, while others may exhibit high CD8+ T cell infiltration but elevated expression of other immune-inhibitory molecules [161].

3.4. Hyperprogression

Hyperprogression, a phenomenon characterised by accelerated tumour growth following treatment with immune checkpoint inhibitors (ICIs), is associated with poor prognosis in lung cancer. Emerging evidence suggests that this rapid disease progression is driven by a complex interplay of genetic alterations, metabolic dysregulation, microbial influences, and epigenetic modifications.

3.4.1. Genetic Mutations

Several genetic mutations have been implicated in hyperprogression by fostering an immunosuppressive tumour microenvironment (TME) and promoting resistance to ICIs:
  • STK11 (LKB1) mutations contribute to an immunosuppressive TME by increasing neutrophil recruitment and proinflammatory cytokine production, leading to poor responses to ICIs and an increased likelihood of hyperprogression [162].
  • KEAP1 mutations drive hyperactivation of the NRF2 pathway, which enhances immune evasion and metabolic reprogramming, ultimately supporting tumour growth and ICI resistance [137].
  • JAK1/2 mutations impair interferon-gamma signalling, disrupting antigen presentation and T-cell activation. This immune dysfunction can facilitate rapid tumour progression during immunotherapy [137].

3.4.2. Metabolic Dysregulation

Metabolic reprogramming in tumours can create an immunosuppressive TME that fosters hyperprogression. For example, increased glycolysis and lactate production, particularly in tumours harbouring STK11 mutations, suppress antitumour immunity, thereby exacerbating resistance to ICIs [163].

3.4.3. Microbial Influences

The gut microbiome plays a crucial role in modulating systemic immune responses and the efficacy of ICIs. Dysbiosis or specific microbial compositions can alter immune cell function and cytokine production, thereby influencing tumour progression and potentially contributing to hyperprogression [163].

3.4.4. Epigenetic Modifications

Epigenetic alterations, including DNA methylation and histone modifications, can suppress immune-related gene expression, further promoting hyperprogression. For instance, epigenetic silencing of interferon-stimulated genes in JAK1/2-mutant tumours has been shown to exacerbate ICI resistance and accelerate tumour progression [137].
By elucidating these molecular mechanisms, ongoing research aims to identify predictive biomarkers and therapeutic strategies to mitigate hyperprogression in lung cancer patients undergoing ICI therapy.

4. Strategies to Overcome Resistance to Immunotherapy

Overcoming immunotherapy resistance represents one of the most significant challenges in lung cancer treatment, particularly in non-small cell lung cancer (NSCLC) [129]. As our understanding of resistance mechanisms continues to evolve, several innovative strategies have emerged to address both primary and acquired resistance, considering factors such as comorbidities, genomic alterations, and changes in the tumour microenvironment (TME).

4.1. Combination Therapies

The development of combination therapies has marked a significant advancement in overcoming immunotherapy resistance. These approaches combine immune checkpoint inhibitors (ICIs) with various treatment modalities, including chemotherapy, targeted therapies, anti-angiogenic agents, and other modern drugs [164]. The future of lung cancer treatment lies in research on novel immunotherapies, both as monotherapies and in innovative drug combinations [165].
Chemotherapy and Immunotherapy Combinations
Chemotherapy-immunotherapy combinations have demonstrated particular efficacy through their ability to trigger immunogenic cell death [166]. The landmark KEYNOTE-189 trial provided compelling evidence for this approach, showing significantly improved survival outcomes when pembrolizumab was combined with chemotherapy in NSCLC [44]. Similarly, studies by Reck et al. demonstrated that atezolizumab, when paired with bevacizumab and chemotherapy, improved progression-free survival and overall survival, regardless of PD-L1 expression and EGFR or ALK genetic alteration status [45].
Targeted Therapy Combinations
In patients with specific molecular alterations, combining targeted therapies with immunotherapy offers a promising strategy, particularly in overcoming resistance associated with long-term targeted therapy use. Studies by Nakagawa et al. have examined the dual blockade of EGFR and VEGF pathways in EGFR-mutated metastatic NSCLC, with phase three trials of EGFR TKI combined with ramucirumab showing positive outcomes on progression-free survival [57].
Combination therapies targeting CTLA-4 (ipilimumab) and PD-1/PD-L1 (nivolumab) have shown improved survival in NSCLC. Ongoing phase 3 trials are investigating coinhibitory targets such as TIGIT, VISTA, LAG3, TIM3, and Siglec-15, where tiragolumab (anti-TIGIT) combined with atezolizumab (anti–PD-L1) has demonstrated increased overall response rate and progression-free survival in PD-L1-positive NSCLC patients [28,102]. Additionally, promising results have emerged from research combining anti-VEGF agents (like bevacizumab or lenvatinib) with ICI [28,45].
Radiation Therapy Combinations
Radiotherapy, traditionally used for local tumour control, has demonstrated significant potential when combined with immunotherapy. For patients with limited progression sites, this combination has shown potential benefits [167], with studies by Theelen et al. demonstrating improved overall survival in NSCLC patients when radiotherapy was combined with pembrolizumab [52]. The synergistic effect extends beyond local control, with some cases demonstrating the abscopal effect, where non-irradiated metastatic sites also show response to treatment.
Novel Combinations and Emerging Approaches
Current research is exploring various pathways involving transforming growth factor β (TGFβ), IL-1β, PARP, VEGF, and cMET [28]. Although initial studies suggested that targeting inflammation (and thus cytokines) may be beneficial in diseases such as cancer, research on IL-1β inhibition (canakinumab) as well as other cytokine targets, like IL-10 and TGF-β, did not demonstrate an overall survival benefit in phase III trials which can be related to several factors [28,168]. This is largely due to the complex role of cytokines in the immune system: some cytokines promote tumour growth, while others are critical for immune surveillance [168]. For example, inhibiting IL-1β can reduce inflammation but can also impair antitumour immune responses [28]. Similarly, IL-10 and TGF-γ play roles in both inhibiting and promoting tumour progression, depending on the context [168]. Ultimately, these therapies have failed to demonstrate consistent clinical benefit in late-phase studies [28,168].
The combination of BRAF and mitogen-activated protein kinase (MEK) inhibitors (dabrafenib and trametinib) has shown efficacy in BRAF V600E-mutant NSCLC, as demonstrated by Planchard et al. [112]. Additionally, therapeutic cancer vaccines, when combined with checkpoint inhibitors, aim to potentiate immune responses against cancer [113].

4.2. Targeting the Tumour Microenvironment

The tumour microenvironment plays a crucial role in immunotherapy resistance, necessitating targeted strategies to modify this complex ecosystem [169]. Current approaches focus on inhibiting immunosuppressive cells and cytokines, modulating tumour metabolism, and enhancing T-cell trafficking, survival, and function [128]. The phase II COAST trial, evaluating the anti-CD73 antibody oleclumab in combination with durvalumab in stage III NSCLC, represents a significant step forward in targeting the TME [170].
Novel intratumoral therapies, including toll-like receptor agonists, STING agonists, and oncolytic viruses, are being investigated to modulate the TME and enhance ICI efficacy [171]. Additionally, nanomedicine delivery systems are being developed to enhance the local concentration of ICIs and reduce systemic side effects [172].

4.3. Addressing Genomic Alterations

The identification of specific genetic mutations driving resistance to immune checkpoint inhibitors (ICIs) has led to the development of targeted therapeutic strategies tailored to distinct molecular profiles. Among these, mutations in STK11, KEAP1, and JAK1/2 have been recognised as key determinants of immune response and treatment outcomes. Current approaches aim to counteract the immunosuppressive effects of these mutations and enhance the efficacy of immunotherapy.
Mutations in STK11 create a non-T cell-inflamed tumour microenvironment (TME), which reduces responsiveness to immune checkpoint inhibitors (ICIs). To overcome this resistance, researchers have explored combination therapies that integrate CTLA-4 inhibitors, such as tremelimumab, to enhance T-cell priming. Additionally, efforts to modulate the inflammatory landscape through IL-6-neutralising or neutrophil-depleting antibodies show promise in reversing the immunosuppressive effects of STK11-driven tumours. Metabolic interventions targeting tumour-associated metabolic reprogramming may further restore immune activity and improve responses to ICIs [140].
Similarly, KEAP1 mutations drive hyperactivation of the NRF2 pathway, promoting immune evasion through metabolic reprogramming. Recent studies highlight the potential of combining glutaminase inhibitors with ICIs to exploit metabolic vulnerabilities in KEAP1-mutant tumours. Other approaches focus on pathway modulation to counteract NRF2-driven immune suppression, offering new avenues for restoring immune sensitivity [136].
In contrast, JAK1/2 mutations disrupt interferon-gamma signalling, impairing antigen presentation and T-cell activation. Strategies aimed at restoring or bypassing interferon signalling pathways have been proposed to enhance immune recognition in these tumours. Therapeutic approaches that improve antigen presentation and bolster T cell activation through combination therapies are also being investigated to counteract this form of resistance [137].
By tailoring treatment strategies to the underlying genetic landscape, these targeted approaches aim to overcome immunotherapy resistance and extend the benefits of ICIs to a broader range of patients. Ongoing research continues to refine these interventions, bringing new hope for more effective and personalised treatments for lung cancer.

4.4. Impact of Comorbidities

Comorbidities, particularly chronic obstructive pulmonary disease (COPD), can significantly influence immune responses and treatment outcomes. Optimising COPD treatment has shown the potential to improve overall immune function and potentially enhance the efficacy of ICIs [28]. Management of comorbidities represents an essential component of comprehensive immunotherapy resistance strategies.

4.5. Novel Biomarkers and Immune Checkpoints

The exploration of new immune checkpoints and biomarkers has emerged as a crucial strategy for overcoming resistance. PD-L1 expression and tumour mutation burden (TMB) remain the most applied biomarkers for predicting immunotherapy response [173], with NSCLC patients showing high PD-L1 expression typically obtaining better benefits from anti-PD-1/PD-L1 therapy [174].
Recent advances include investigation of novel checkpoint inhibitors targeting LAG-3, TIM-3, and other proteins. Relatlimab, a LAG-3 inhibitor, combined with nivolumab has shown promising activity in PD-L1-positive NSCLC [114]. Similarly, sabatolimab, an anti-TIM-3 IgG4 monoclonal antibody, has demonstrated potential in combination studies [26,116].
TIGIT blockade has shown particular promise, with tiragolumab combination therapy significantly enhancing progression-free survival and overall response rate in PD-L1-positive patients [117]. The development of ICOS (Inducible T-Cell Co-Stimulator) targeting approaches, such as GSK3359609, has also shown encouraging early results [120].

4.6. Liquid Biopsy for Detecting ICI Sensitivity and Resistance Biomarkers

Liquid biopsy is an emerging, minimally invasive technique that analyses circulating tumour-derived components in bodily fluids, offering significant potential in oncology for detecting biomarkers associated with sensitivity and resistance to immune checkpoint inhibitors (ICIs). Key analytes in liquid biopsies include circulating tumour DNA (ctDNA), circulating tumour cells (CTCs), and extracellular vesicles, which provide insights into the tumour’s genetic and epigenetic landscape [175,176].
Liquid biopsies can identify biomarkers predictive of a positive response to ICIs. For instance, tumour mutational burden (TMB) can be assessed through ctDNA analysis, providing a non-invasive means to predict ICI efficacy. High TMB levels have been associated with better responses to ICIs across various cancers. Additionally, monitoring dynamic changes in ctDNA levels during treatment can offer real-time insights into therapeutic responses [177].
Liquid biopsies are instrumental in identifying mechanisms of resistance to ICIs. For example, specific genetic alterations detected in ctDNA, such as mutations in the JAK1/2 genes, have been linked to acquired resistance to PD-1 blockade therapies. Furthermore, the detection of CTCs expressing immune checkpoint ligands like PD-L1 can provide insights into tumour immune evasion strategies. The primary advantage of liquid biopsy lies in its ability to provide a comprehensive and dynamic overview of tumour heterogeneity and evolution, which is often challenging with traditional tissue biopsies. However, challenges remain, including standardising methodologies, ensuring analytical sensitivity and specificity, and validating findings across large patient cohorts. Despite these hurdles, liquid biopsy holds promise for guiding immunotherapy decisions and monitoring treatment responses in real time [178,179,180].

4.7. Adaptive Clinical Trial Designs

Adaptive clinical trial design can significantly help in overcoming resistance to immunotherapy in lung cancer by allowing for real-time modifications based on interim data, thereby addressing the roles of metabolic, microbial, and epigenetic factors, as well as genetic mutations.
Adaptive trials can incorporate biomarker-driven strategies to tailor treatments to individual patient profiles. For instance, patients with STK11 mutations, which are associated with an immunosuppressive tumour microenvironment (TME), can be stratified and treated with combination therapies that include immune checkpoint inhibitors (ICIs) and metabolic modulators to counteract the metabolic reprogramming that promotes resistance [181,182].
For KEAP1 mutations, which lead to hyperactivation of the NRF2 pathway and immune evasion, adaptive trials can evaluate the efficacy of combining ICIs with glutaminase inhibitors. This approach targets the metabolic vulnerabilities of KEAP1-mutant tumours, potentially enhancing the response to immunotherapy [181,183].
JAK1/2 mutations disrupt interferon-gamma signalling, impairing antigen presentation and T cell activation. Adaptive trial designs can facilitate the testing of agents that restore or bypass these pathways, such as epigenetic therapies that enhance antigen presentation or T cell activation, thereby improving outcomes in JAK1/2-mutant tumours [139,181].
Additionally, adaptive trials can incorporate microbial factors by monitoring the gut microbiome and adjusting treatments based on microbiome composition, which has been shown to influence the efficacy of ICIs [181,182].
By using master protocols and biomarker-adaptive randomization, adaptive trials can efficiently identify and validate the most effective treatment combinations for overcoming resistance, ultimately improving overall survival (OS) and progression-free survival (PFS) in lung cancer patients [184,185,186].

4.8. Recent Clinical Trials Targeting Resistance to Immunotherapy in Lung Cancer

Several recent clinical trials are targeting resistance to immunotherapy in lung cancer, particularly for patients with non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC), considering factors such as comorbidities like chronic obstructive pulmonary disease (COPD), genomic alterations, and changes in the tumour microenvironment (TME). They are summarised in Table 5.

5. Future Directions

Despite recent progress in precision medicine and numerous ongoing clinical trials, several critical challenges and questions remain to be addressed, namely:

5.1. Treatment Response and Resistance

First of all, immunotherapy response rates still remain relatively low. Current combination therapies, while more effective than single agents, still achieve response rates below 50%. PD1/L1 expression alone has proven insufficient as a predictor of response. Even among responding patients, complete tumour elimination is rare, even if occurs, especially in bladder and melanoma cancer patients. Remaining cells may lead to relapse, often developing different immunosuppression mechanisms and mutations compared to the original tumour.
Probably treatment optimization might be the key, apart from other factors, such as biomarkers. The optimal combination and sequencing of targeted therapy, radiotherapy, chemotherapy, and immunotherapy remain unclear, despite many ongoing trials. Questions persist about the timing of immunotherapy, particularly regarding the patient’s immune system status after chemotherapy. Finally, the role of neoadjuvant chemotherapy requires further investigation.

5.2. Biological Understanding and Patient Factors

Our understanding of cancer biology remains in its infancy. The durability of responses to immunotherapy likely connects to biological mechanisms that are yet to be discovered, requiring further basic science research to understand the complex pathways involved. Moreover, adverse effects and the hyperprogression phenomenon also require more attention, particularly regarding the variability in adverse effect severity among patients. Some patients develop aggressive autoimmune progression or hyperprogression despite initially having very good prognostic biomarkers. Factors involved may include immune system function, organism-wide effects, or unidentified environmental influences.
Finally, regarding gender and racial differences: research has revealed significant variations between males and females in gene expression and methylation status, suggesting the need for gender-specific treatment approaches. This issue has been recently raised by many international committees and, as a result, more clinical trials will encompass inclusiveness or will be conducted separately for specific ethnic groups.

5.3. Future Research Priorities

The path forward requires a coordinated effort from the scientific community, combining basic research with clinical applications. Integration of multi-omics approaches, and advanced modelling techniques will be essential for unravelling the complexities of cancer biology and improving patient outcomes. Success will depend on developing personalised, multi-modal approaches tailored to each patient’s unique tumour and immune profile, supported by adaptive trial designs and biomarker-driven treatment selection.
The most important research directions for lung immunotherapy have been presented in bullet points below:
  • Biomarker Development:
    • urgent need for reliable biomarker detection,
    • potential of liquid biopsy techniques using circulating tumour DNA,
    • development of markers for tumour presence, progression, metastases, and treatment response,
    • identification of markers to predict treatment benefits and adverse effect risks.
  • Treatment Optimization:
    • precise dosage administration,
    • expansion of pharmacogenetics and pharmacogenomics,
    • implementation of genetic testing before drug administration,
    • standardization of resistance definitions,
    • development of comprehensive genomic profiling with immune gene signatures.
  • Novel Therapeutic Approaches:
    • personalised cancer vaccine development,
    • investigation of epigenetic modulation,
    • metabolic manipulation strategies,
    • gut microbiome modulation,
    • exploration of immunotherapy rechallenge in previously responsive patients.
  • Neoantigen Research:
    • further investigation into the stochastic nature of neoantigen expression and its targeting potential.

5.4. Future Directions and Remaining Open Questions

In summary, despite recent advances in precision medicine and numerous ongoing clinical trials, several critical questions remain unresolved. We propose addressing these challenges collectively as a scientific community:
  • The limited response rate to immunotherapy remains problematic. Although combination therapies demonstrate enhanced efficacy, response rates rarely exceed 50% and are typically substantially lower. PD-1/PD-L1 expression alone proves insufficient as a predictive biomarker for potential responders.
  • Even among responsive patients, complete tumour cell elimination is rare, with residual cells potentially facilitating future relapse through the development of resistant clones. Such recurrent tumours frequently exhibit altered immunosuppressive mechanisms and mutational profiles distinct from their progenitor lesions. Consequently, the optimization of first-line therapy, specifically tailored to individual patients, remains crucial.
  • The optimal integration of targeted therapy, radiotherapy, chemotherapy, and/or immunotherapy remains undefined. Similarly, the appropriate therapeutic sequence requires clarification, as preliminary data suggest immunotherapy efficacy may be compromised in patients who have undergone chemotherapy due to immune system exhaustion or suppression. The role of neoadjuvant chemotherapy, in particular, warrants further investigation.
  • When effective, immunotherapeutic responses demonstrate remarkable durability. However, the biological mechanisms underlying this sustained response remain poorly elucidated and necessitate fundamental scientific investigation. Enhanced comprehension of cancer biology will facilitate more precise targeting of critical nodes within complex pathways.
  • The heterogeneity in adverse event severity among patients receiving immunotherapy remains unexplained. This variability may be attributable to intrinsic immune system function, systemic physiological factors, or unidentified environmental variables that have yet to be characterised.
  • The etiology of hyperprogressions and severe autoimmune complications following immunotherapy administration remains unclear. A subset of patients experiences rapid deterioration and mortality shortly after immune checkpoint inhibitor initiation. Current knowledge is insufficient to explain this phenomenon adequately or to implement appropriate interventional strategies.
  • While it is now widely accepted that tumours express targetable neoantigens, their expression patterns appear stochastic, necessitating substantial additional research in this domain.
  • Therapeutic dosage optimization requires precision, yet sufficient data to accomplish this effectively remains lacking. Pharmacogenetics and pharmacogenomics offer promising approaches, though these analyses are not universally implemented. In many regions, they remain insufficiently adopted or are employed reactively following adverse events rather than proactively before treatment initiation.
  • There exists an urgent requirement for robust biomarker identification. Emerging diagnostic technologies based on circulating tumour DNA (ctDNA)—termed liquid biopsy—hold significant potential for identifying molecules that serve as biomarkers for tumour presence, progression, metastasis, treatment selection, and therapeutic response monitoring. Specific biomarkers would also facilitate the identification of patients most likely to benefit from therapy while minimising adverse effects.
  • Multiple investigations have demonstrated significant sex-based differences in gene expression and methylation status. These findings suggest that therapeutic approaches should be further refined according to patients’ biological sex among other relevant factors. In oncology, precision medicine represents a necessity rather than an optional approach for clinicians or institutions.
  • Although numerous factors are known to influence patient response to immunotherapy, our limited understanding of underlying mechanisms limits their clinical application. For instance, low-light conditions are known to induce a more inflammatory microenvironment, potentially enhancing immunotherapeutic efficacy while possibly compromising mental health. Similar considerations apply to caloric restriction.

5.5. Insights from the Genomic Medicine Perspective

Genetic variants may influence the prediction of the treatment of lung cancer. For example, a set of 140 genes may predict the response and survival to anti-PD-L1 therapy in non-small cell lung cancer. The set of genes may help to identify 5 classes of clinically aggressive tumours that can respond to immunotherapy. In turn, in the case of lung adenocarcinoma, a set of 10 genes and their genetic variants associated with chemotherapy resistance has been identified (namely, PLEK2, TFAP2A, KIF20A, S100P, GDF15, HSPB8, SASH1, WASF3, LAMA3 and TCN1). Further, a subset of genes related to a particular clinical outcome may be delineated. For example, TP53 alterations and co-alterations of TP53 missense mutations with TP73, CREBBP/EP300 or FMN2 are indicative of a shorter disease prolapse after chemotherapy and some genetic patterns associated with resistance to chemotherapy [193]. The most important genetic insights have been summarised in Table 6. Furthermore, deep learning algorithms may identify expression profiles related to the metastatic potential of the tumour [194].

6. Conclusions

Lung cancer remains an example of malignancy with a very poor prognosis despite the recent progress in medicine. Different treatment strategies are now available for cancer therapy based on its type, molecular subtype and other factors including overall health, the stage of disease and cancer molecular profile. Immunotherapy is emerging as a potential addition to surgery, chemotherapy, radiotherapy or other targeted therapies, but also considered a mainstay therapy mode. In fact, the pace of research and clinical studies on cancer immunotherapy seem to outride the progress in understanding the biological background underlying behind. The lung cancer microenvironment is a complex and dynamic entity that significantly influences tumour behaviour and therapeutic response. Continued research into the cellular and molecular mechanisms within the TME will provide insights into novel therapeutic targets and strategies. Integrating multi-omics approaches and advanced modelling techniques will be essential for unravelling the intricacies of the TME and improving outcomes for lung cancer patients [195,196].

Funding

This research received no external funding.

Data Availability Statement

This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ADAMTSmetalloproteinase with thrombospondin motifs
ALKanaplastic lymphoma kinase
CAFscancer-associated fibroblasts
COPDchronic obstructive pulmonary disease
ctDNAcirculating tumour DNA
CTLA-4cytotoxic T cell antigen 4
DNAdeoxyribonucleic acid
ECMextracellular matrix
EGFRepidermal growth factor receptor
EMAEuropean Medicines Agency
EUEuropean Union
FDAFood and Drug Administration
FGF-2fibroblast growth factor-2
HDAChistone deacetylase
HIFhypoxia-inducible factors
ICIimmune checkpoint inhibitor
ICOSLinducible T-cell costimulator ligand
IFNinterferon
KEAPKelch-like ECH-associated protein 1
KRAS geneKirsten rat sarcoma gene
LClung cancer
MDSCmyeloid-derived suppressor cells
MEKmitogen-activated protein kinase
METmesenchymal epithelial transition receptor
MMPsmatrix metalloproteinases
NKnatural killer cells
NSCLCnon-small cell lung cancer
NTRKneurotrophic tyrosine receptor kinase
PD-1/PD-L1programmed death protein-1
PVRpoliovirus receptor
RNAribonucleic acid
SCLCsmall cell lung cancer
TGFTransforming growth factor
TKItyrosine kinase inhibitors
TMEtumour microenvironment
VEGFvascular endothelial growth factor
VEGFR2vascular endothelial growth factor receptor 2

References

  1. Modlińska, A.; Kowalczyk, A. Lung Cancer—Epidemiology, Clinical Symptoms and Social Consequences. Psychoonkologia 2016, 20, 57–65. [Google Scholar] [CrossRef]
  2. Islami, F.; Torre, L.A.; Jemal, A. Global Trends of Lung Cancer Mortality and Smoking Prevalence. Transl. Lung Cancer Res. 2015, 4, 327–338. [Google Scholar] [PubMed]
  3. Ma, W.; Gilligan, B.M.; Yuan, J.; Li, T. Current Status and Perspectives in Translational Biomarker Research for PD-1/PD-L1 Immune Checkpoint Blockade Therapy. J. Hematol. Oncol. 2016, 9, 47. [Google Scholar] [CrossRef] [PubMed]
  4. Mahoney, K.M.; Rennert, P.D.; Freeman, G.J. Combination Cancer Immunotherapy and New Immunomodulatory targets. Nat. Rev. Drug Discov. 2015, 14, 561–584. [Google Scholar] [CrossRef]
  5. Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman Jeffrey, A.; Atkins, M.B.; et al. Safety, Activity, and Immune Correlates of Anti-PD-1 antibody in Cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef]
  6. Shiravand, Y.; Khodadadi, F.; Kashani, S.M.A.; Hosseini-Fard, S.R.; Hosseini, S.; Sadeghirad, H.; Ladwa, R.; O’Byrne, K.; Kulasinghe, A. Immune Checkpoint Inhibitors in Cancer Therapy. Curr. Oncol. 2022, 29, 3044–3060. [Google Scholar] [CrossRef] [PubMed]
  7. McGranahan, N.; Furness, A.J.S.; Rosenthal, R.; Ramskov, S.; Lyngaa, R.; Saini, S.K.; Jamal-Hanjani, M.; Wilson, G.A.; Birkbak, N.J.; Hiley, C.T.; et al. Clonal Neoantigens Elicit T Cell Immunoreactivity and sensitivity to Immune Checkpoint Blockade. Science (1979) 2016, 351, 1463–1469. [Google Scholar] [CrossRef]
  8. Fukuda, S.; Suda, K.; Hamada, A.; Tsutani, Y. Recent Advances in Perioperative Immunotherapies in Lung Cancer. Biomolecules 2023, 13, 1377. [Google Scholar] [CrossRef]
  9. Ren, Z.; Shang, S.; Chen, D. Recent Advances in Immunotherapy for Small Cell Lung Cancer. Curr. Opin. Oncol. 2025, 37, 17–26. [Google Scholar] [CrossRef]
  10. Dobosz, P.; Stępień, M.; Golke, A.; Dzieciątkowski, T. Challenges of the Immunotherapy: Perspectives and Limitations of the Immune Checkpoint Inhibitor Treatment. Int. J. Mol. Sci. 2022, 23, 2847. [Google Scholar] [CrossRef]
  11. Putzu, C.; Canova, S.; Paliogiannis, P.; Lobrano, R.; Sala, L.; Cortinovis, D.L.; Colonese, F. Duration of Immunotherapy in Non-Small Cell Lung Cancer Survivors: A Lifelong Commitment? Cancers 2023, 15, 689. [Google Scholar] [CrossRef] [PubMed]
  12. Chen, D.S.; Mellman, I. Elements of Cancer Immunity and the Cancer–Immune Set Point. Nature 2017, 541, 321–330. [Google Scholar] [CrossRef] [PubMed]
  13. Turnis, M.E.; Andrews, L.P.; Vignali, D.A.A. Inhibitory Receptors as Targets for Cancer Immunotherapy. Eur. J. Immunol. 2015, 45, 1892–1905. [Google Scholar] [CrossRef]
  14. Samstein, R.M.; Lee, C.-H.; Shoushtari, A.N.; Hellmann, M.D.; Shen, R.; Janjigian, Y.Y.; Barron, D.A.; Zehir, A.; Jordan, E.J.; Omuro, A.; et al. Tumor Mutational Load Predicts Survival after immunotherapy across Multiple Cancer Types. Nat. Genet. 2019, 51, 202–206. [Google Scholar] [CrossRef] [PubMed]
  15. Zhou, C.; Li, M.; Wang, Z.; An, D.; Li, B. Adverse Events of Immunotherapy in Non-Small Cell Lung Cancer: A Review and Network Meta-Analysis. Int. Immunopharmacol. 2022, 102, 108353. [Google Scholar] [CrossRef]
  16. O’Donnell, J.S.; Long, G.V.; Scolyer, R.A.; Teng, M.W.L.; Smyth, M.J. Resistance to PD1/PDL1 Checkpoint Inhibition. Cancer Treat. Rev. 2017, 52, 71–81. [Google Scholar] [CrossRef]
  17. Rapoport, B.L.; Shannon, V.R.; Cooksley, T.; Johnson, D.B.; Anderson, L.; Blidner, A.G.; Tintinger, G.R.; Anderson, R. Pulmonary Toxicities Associated with the Use of Immune Checkpoint Inhibitors: An Update from the Immuno-Oncology Subgroup of the Neutropenia, Infection & Myelosuppression Study Group of the Multinational Association for Supportive Care in Cancer. Front. Pharmacol. 2021, 12, 743582. [Google Scholar]
  18. Shah, D.; Soper, B.; Shopland, L. Cytokine Release Syndrome and Cancer Immunotherapies—Historical and Promising Futures. Front. Immunol. 2023, 14, 1190379. [Google Scholar] [CrossRef]
  19. Khalil, D.N.; Smith, E.L.; Brentjens, R.J.; Wolchok, J.D. The Future of Cancer Treatment: Immunomodulation, CARs and combination Immunotherapy. Nat. Rev. Clin. Oncol. 2016, 13, 273–290. [Google Scholar] [CrossRef]
  20. Adashek, J.J.; Subbiah, I.M.; Matos, I.; Garralda, E.; Menta, A.K.; Ganeshan, D.M.; Subbiah, V. Hyperprogression and Immunotherapy: Fact, Fiction, or alternative Fact? Trends Cancer 2020, 6, 181–191. [Google Scholar] [CrossRef]
  21. Sehgal, K. Hyperprogression in Patients with Cancer Receiving Immune Checkpoint Inhibitors. JAMA Netw. Open 2021, 4, e211839. [Google Scholar] [CrossRef] [PubMed]
  22. Britt, A.S.; Huang, C.; Huang, C.H. Hyperprogressive Disease in Non-Small Cell Lung Cancer treated with Immune Checkpoint Inhibitor Therapy, Fact or Myth? Front. Oncol. 2022, 12, 996554. [Google Scholar] [CrossRef] [PubMed]
  23. Dobosz, P.; Stempor, P.A.; Roszik, J.; Herman, A.; Layani, A.; Berger, R.; Avni, D.; Sidi, Y.; Leibowitz-Amit, R. Checkpoint Genes at the Cancer Side of the Immunological synapse in Bladder Cancer. Transl. Oncol. 2020, 13, 193–200. [Google Scholar] [CrossRef]
  24. Stempor, P.A.; Avni, D.; Leibowitz, R.; Sidi, Y.; Stepień, M.; Dzieciatkowski, T.; Dobosz, P. Comprehensive Analysis of Correlations in the Expression of miRNA Genes and Immune Checkpoint Genes in Bladder Cancer. Int. J. Mol. Sci. 2021, 22, 2553. [Google Scholar] [CrossRef]
  25. Liu, W.-F.; Quan, B.; Li, M.; Zhang, F.; Hu, K.-S.; Yin, X. PVR-A Prognostic Biomarker Correlated with Immune Cell in Hepatocellular Carcinoma. Diagnostics 2022, 12, 2953. [Google Scholar] [CrossRef]
  26. Shen, Y.; Chen, J.; Li, X.-P. Research Advances in Immune Checkpoint Drugs for Non-Small Cell Lung Cancer. J. Drug Target. 2023, 31, 700–713. [Google Scholar] [CrossRef] [PubMed]
  27. Lahiri, A.; Maji, A.; Potdar, P.D.; Singh, N.; Parikh, P.; Bisht, B.; Mukherjee, A.; Paul, M.K. Lung Cancer Immunotherapy: Progress, Pitfalls, and Promises. Mol. Cancer 2023, 22, 40. [Google Scholar] [CrossRef]
  28. 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]
  29. 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]
  30. Kim, S.I.; Cassella, C.R.; Byrne, K.T. Tumor Burden and Immunotherapy: Impact on Immune Infiltration and Therapeutic Outcomes. Front. Immunol. 2021, 11, 629722. [Google Scholar] [CrossRef]
  31. Lieber, A.; Makai, A.; Orosz, Z.; Kardos, T.; Isaac, S.J.; Tornyi, I.; Bittner, N. The Role of Immunotherapy in Early-Stage and Metastatic NSCLC. Pathol. Oncol. Res. 2024, 30, 1611713. [Google Scholar] [CrossRef] [PubMed]
  32. Doroudian, M.; Zanganeh, S.; Abbasgholinejad, E.; Donnelly, S.C. Nanomedicine in Lung Cancer Immunotherapy. Front. Bioeng. Biotechnol. 2023, 11, 1144653. [Google Scholar] [CrossRef] [PubMed]
  33. 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]
  34. Sun, Y.; Yang, Q.; Shen, J.; Wei, T.; Shen, W.; Zhang, N.; Luo, P.; Zhang, J. The Effect of Smoking on the Immune Microenvironment and Immunogenicity and Its Relationship With the Prognosis of Immune Checkpoint Inhibitors in Non-Small Cell Lung Cancer. Front. Cell Dev. Biol. 2021, 9, 745859. [Google Scholar] [CrossRef]
  35. Yin, Q.; Wu, L.; Han, L.; Zheng, X.; Tong, R.; Li, L.; Bai, L.; Bian, Y. Immune-Related Adverse Events of Immune Checkpoint Inhibitors: A Review. Front. Immunol. 2023, 14, 1167975. [Google Scholar] [CrossRef]
  36. Predina, J.; Eruslanov, E.; Judy, B.; Kapoor, V.; Cheng, G.; Wang, L.-C.; Sun, J.; Moon, E.K.; Fridlender, Z.G.; Albelda, S.; et al. Changes in the Local Tumor Microenvironment in Recurrent Cancers may Explain the Failure of Vaccines after Surgery. Proc. Natl. Acad. Sci. USA 2013, 110, E415–E424. [Google Scholar] [CrossRef]
  37. Huynh, C.; Walsh, L.A.; Spicer, J.D. Surgery after Neoadjuvant Immunotherapy in Patients with resectable Non-Small Cell Lung Cancer. Transl. Lung Cancer Res. 2021, 10, 563–580. [Google Scholar] [CrossRef]
  38. Forget, P.; Simonet, O.; De Kock, M. Cancer Surgery Induces Inflammation, Immunosuppression and neo-Angiogenesis, but Is It Influenced by Analgesics? F1000Res 2013, 2, 102. [Google Scholar] [CrossRef] [PubMed]
  39. Carbone, D.P.; Gandara, D.R.; Antonia, S.J.; Zielinski, C.; Paz-Ares, L. Non-Small-Cell Lung Cancer: Role of the Immune System and potential for Immunotherapy. J. Thorac. Oncol. 2015, 10, 974–984. [Google Scholar] [CrossRef]
  40. Rami-Porta, R. The Evolving Concept of Complete Resection in Lung Cancer Surgery. Cancers 2021, 13, 2583. [Google Scholar] [CrossRef]
  41. Wong, L.-Y.; Dale, R.; Kapula, N.; Elliott, I.A.; Liou, D.Z.; Backhus, L.M.; Lui, N.S.; Shrager, J.B.; Berry, M.F. Impacts of Positive Margins and Surgical Extent on outcomes after Early-Stage Lung Cancer Resection. Ann. Thorac. Surg. 2024, 118, 1126–1134. [Google Scholar] [CrossRef]
  42. Lee, S.H. Chemotherapy for Lung Cancer in the Era of Personalized Medicine. Tuberc. Respir. Dis. 2019, 82, 179. [Google Scholar] [CrossRef] [PubMed]
  43. Li, J.-Y.; Chen, Y.-P.; Li, Y.-Q.; Liu, N.; Ma, J. Chemotherapeutic and Targeted Agents Can Modulate the Tumor Microenvironment and Increase the Efficacy of Immune Checkpoint Blockades. Mol. Cancer 2021, 20, 27. [Google Scholar] [CrossRef]
  44. Gandhi, L.; Rodriguez-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 Cancer. N. Engl. J. Med. 2018, 378, 2078–2092. [Google Scholar] [CrossRef] [PubMed]
  45. 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] [PubMed]
  46. Fendler, W.; Tomasik, B.; Atkins, K.; Stawiski, K.; Chałubińska-Fendler, J.; Kozono, D. The Clinician’s Guide to Radiotherapy Complications. Pol. Arch. Intern. Med. 2022, 132, 1–11. [Google Scholar] [CrossRef]
  47. Zhang, Z.; Peng, Y.; Peng, X.; Xiao, D.; Shi, Y.; Tao, Y. Effects of Radiation Therapy on Tumor Microenvironment: An Updated Review. Chin. Med. J. 2023, 136, 2802–2811. [Google Scholar] [CrossRef]
  48. Vignard, J.; Mirey, G.; Salles, B. Ionizing-Radiation Induced DNA Double-Strand Breaks: A Direct and Indirect Lighting Up. Radiother. Oncol. 2013, 108, 362–369. [Google Scholar] [CrossRef]
  49. Deng, L.; Liang, H.; Xu, M.; Yang, X.; Burnette, B.; Arina, A.; Li, X.-D.; Mauceri, H.; Beckett, M.; Darga, T.; et al. STING-Dependent Cytosolic DNA Sensing Promotes Radiation-Induced Type I Interferon-Dependent Antitumor Immunity in Immunogenic Tumors. Immunity 2014, 41, 843–852. [Google Scholar] [CrossRef]
  50. Simon Davis, D.A.; Atmosukarto, I.I.; Garrett, J.; Gosling, K.; Syed, F.M.; Quah, B.J. Irradiation Immunity Interactions. J. Med. Imaging Radiat. Oncol. 2022, 66, 519–535. [Google Scholar] [CrossRef]
  51. Huang, R.-X.; Zhou, P.-K. DNA Damage Response Signaling Pathways and Targets for radiotherapy Sensitization in Cancer. Signal Transduct. Target. Ther. 2020, 5, 60. [Google Scholar] [PubMed]
  52. Theelen, W.S.M.E.; Chen, D.; Verma, V.; Hobbs, B.P.; Peulen, H.M.U.; Aerts, J.G.J.V.; Bahce, I.; Niemeijer, A.L.N.; Chang, J.Y.; de Groot, P.M.; et al. Pembrolizumab with or without Radiotherapy for Metastatic Non-Small-Cell Lung Cancer: A Pooled Analysis of Two Randomised Trials. Lancet Respir. Med. 2021, 9, 467–475. [Google Scholar] [CrossRef] [PubMed]
  53. Mayekar, M.K.; Bivona, T.G. Current Landscape of Targeted Therapy in Lung Cancer. Clin. Pharmacol. Ther. 2017, 102, 757–764. [Google Scholar] [CrossRef]
  54. Sekido, Y.; Fong, K.M.; Minna, J.D. Molecular Genetics of Lung Cancer. Annu. Rev. Med. 2003, 54, 73–87. [Google Scholar] [CrossRef]
  55. Li, J.W.; Zheng, G.; Kaye, F.J.; Wu, L. PROTAC Therapy as a New Targeted Therapy for Lung Cancer. Mol. Ther. 2023, 31, 647–656. [Google Scholar] [CrossRef] [PubMed]
  56. Jänne, P.A.; Baik, C.; Su, W.-C.; Johnson, M.L.; Hayashi, H.; Nishio, M.; Kim, D.-W.; Koczywas, M.; Gold, K.A.; Steuer, C.E.; et al. Efficacy and Safety of Patritumab Deruxtecan (HER3-DXd) in EGFR Inhibitor-Resistant, EGFR-Mutated Non-Small Cell Lung Cancer. Cancer Discov. 2022, 12, 74–89. [Google Scholar] [CrossRef]
  57. Nakagawa, K.; Garon, E.B.; Seto, T.; Nishio, M.; Aix, S.P.; Paz-Ares, L.; Chiu, C.; Park, K.; Novello, S.; Nadal, E.; et al. Ramucirumab plus Erlotinib in Patients with Untreated, EGFR-Mutated, Advanced Non-Small-Cell Lung Cancer (RELAY): A, Double-Blind, Placebo-Controlled, Phase 3 Trial. Lancet Oncol. 2019, 20, 1655–1669. [Google Scholar] [CrossRef]
  58. Lin, J.J.; Muzikansky, A.; Kennedy, E.; Kuberski, H.; Stober, L.L.; Wanat, A.C.; Azzoli, C.G.; Lennes, I.; Sequist, L.V.; Dagogo-Jack, I.; et al. Safety and Activity of Alectinib plus Bevacizumab in patients with Advanced ALK-Rearranged Non-Small-Cell Lung Cancer: A Phase I/II Study. ESMO Open 2022, 7, 100342. [Google Scholar] [CrossRef]
  59. Adamek, M.; Biernat, W.; Chorostowska-Wynimko, J.; Didkowska, J.A.; Dziadziuszko, K.; Grodzki, T.; Jassem, J.; Kepka, L.; Kowalski, D.; Krawczyk, P.; et al. Lung Cancer in Poland. J. Thorac. Oncol. 2020, 15, 1271–1276. [Google Scholar] [CrossRef]
  60. Liu, J.; Xue, L.; Xu, X.; Luo, J.; Zhang, S. FAK-Targeting PROTAC Demonstrates Enhanced Antitumor activity against KRAS Mutant Non-Small Cell Lung Cancer. Exp. Cell Res. 2021, 408, 112868. [Google Scholar] [CrossRef]
  61. Bond, M.J.; Chu, L.; Nalawansha, D.A.; Li, K.; Crews, C.M. Targeted Degradation of Oncogenic KRASG12C by VHL-Recruiting. ACS Cent. Sci. 2020, 6, 1367–1375. [Google Scholar] [CrossRef] [PubMed]
  62. Larsen, J.E.; Minna, J.D. Molecular Biology of Lung Cancer: Clinical Implications. Clin. Chest Med. 2011, 32, 703–740. [Google Scholar] [PubMed]
  63. Tada, H.; Mitsudomi, T.; Misumi, T.; Sugio, K.; Tsuboi, M.; Okamoto, I.; Iwamoto, Y.; Sakakura, N.; Sugawara, S.; Atagi, S.; et al. Randomized Phase III Study of Gefitinib versus Cisplatin plus vinorelbine for Patients with Resected Stage II-IIIA Non-Small-Cell Lung Cancer with EGFR Mutation (IMPACT). J. Clin. Oncol. 2022, 40, 231–241. [Google Scholar] [CrossRef] [PubMed]
  64. Hosomi, Y.; Morita, S.; Sugawara, S.; Kato, T.; Fukuhara, T.; Gemma, A.; Takahashi, K.; Fujita, Y.; Harada, T.; Minato, K.; et al. Gefitinib Alone versus Gefitinib plus Chemotherapy For-Small-Cell Lung Cancer with Mutated Epidermal Growth Factor Receptor: NEJ009 Study. J. Clin. Oncol. 2020, 38, 115–123. [Google Scholar] [CrossRef]
  65. Bareschino, M.A.; Schettino, C.; Troiani, T.; Martinelli, E.; Morgillo, F.; Ciardiello, F. Erlotinib in Cancer Treatment. Ann. Oncol. 2007, 18 (Suppl. 6), vi35–vi41. [Google Scholar] [CrossRef]
  66. Perez-Soler, R. The Role of Erlotinib (Tarceva, OSI 774) in the Treatment of non-Small Cell Lung Cancer. Clin. Cancer Res. 2004, 10, 4238s–4240s. [Google Scholar] [CrossRef]
  67. Radonic, T.; Geurts-Giele, W.R.R.; Dubbink, H.J. Response to: RET Fluorescence In Situ Hybridization Analysis Is a Sensitive but Highly Unspecific Screening Method for RET Fusions in Lung Cancer. J. Thorac. Oncol. 2021, 16, e56. [Google Scholar] [CrossRef]
  68. Santarpia, M.; Liguori, A.; Karachaliou, N.; Gonzalez-Cao, M.; Daffinà, M.G.; D’Aveni, A.; Marabello, G.; Altavilla, G.; Rosell, R. Osimertinib in the Treatment of Non-Small-Cell Lung Cancer: Design, Development and Place in Therapy. Lung Cancer (Auckl.) 2017, 8, 109–125. [Google Scholar] [CrossRef]
  69. Carlisle, J.W.; Ramalingam, S.S. Role of Osimertinib in the Treatment of EGFR-Mutation Positive Non-Small-Cell Lung Cancer. Future Oncol. 2019, 15, 805–816. [Google Scholar] [CrossRef]
  70. Wu, Y.-L.; Tsuboi, M.; He, J.; John, T.; Grohe, C.; Majem, M.; Goldman, J.W.; Laktionov, K.; Kim, S.-W.; Kato, T.; et al. Osimertinib in Resected EGFR-Mutated Non-Small-Cell Lung. N. Engl. J. Med. 2020, 383, 1711–1723. [Google Scholar] [CrossRef]
  71. Karachaliou, N.; Fernandez Bruno, M.; Bracht, J.W.P.; Rosell, R. Profile of Alectinib for the Treatment of ALK-Positive Non-Small Cell Lung Cancer (NSCLC): Patient Selection and Perspectives. Onco. Targets. Ther. 2019, 12, 4567–4575. [Google Scholar] [CrossRef]
  72. Peters, S.; Camidge, D.R.; Shaw, A.T.; Gadgeel, S.; Ahn, J.S.; Kim, D.-W.; Ou, S.-H.I.; Pérol, M.; Dziadziuszko, R.; Rosell, R.; et al. Alectinib versus Crizotinib in Untreated ALK-Positive–Small-Cell Lung Cancer. N. Engl. J. Med. 2017, 377, 829–838. [Google Scholar] [CrossRef]
  73. Bedi, S.; Khan, S.A.; AbuKhader, M.M.; Alam, P.; Siddiqui, N.A.; Husain, A. A Comprehensive Review on Brigatinib—A Wonder Drug for targeted Cancer Therapy in Non-Small Cell Lung Cancer. Saudi Pharm. J. 2018, 26, 755–763. [Google Scholar] [CrossRef]
  74. Popat, S.; Brustugun, O.T.; Cadranel, J.; Felip, E.; Garassino, M.C.; Griesinger, F.; Helland, Å.; Hochmair, M.; Pérol, M.; Bent-Ennakhil, N.; et al. Real-World Treatment Outcomes with Brigatinib in Patients with pretreated ALK+ Metastatic Non-Small Cell Lung Cancer. Lung Cancer 2021, 157, 9–16. [Google Scholar] [CrossRef] [PubMed]
  75. Forde, P.M.; Rudin, C.M. Crizotinib in the Treatment of Non-Small-Cell Lung Cancer. Expert. Opin. Pharmacother. 2012, 13, 1195–1201. [Google Scholar] [CrossRef] [PubMed]
  76. Horn, L.; Wang, Z.; Wu, G.; Poddubskaya, E.; Mok, T.; Reck, M.; Wakelee, H.; Chiappori, A.A.; Lee, D.H.; Breder, V.; et al. Ensartinib vs Crizotinib for Patients with Anaplastic Lymphoma-Positive Non-Small Cell Lung Cancer: A Randomized Trial. JAMA Oncol. 2021, 7, 1617–1625. [Google Scholar] [CrossRef] [PubMed]
  77. Niyibizi, B.A.; Muhizi, E.; Ndoli, D.A.; Rukundo, I.; Muvunyi, T.Z.; Musoni, M.; Dukundane, D.; Rudakemwa, E.; Rubagumya, F.; Van Christ Manirakiza, A. Lung Cancer in Rwanda. J. Thorac. Oncol. 2022, 17, 1074–1077. [Google Scholar] [CrossRef]
  78. Solomon, B.J.; Besse, B.; Bauer, T.M.; Felip, E.; Soo, R.A.; Camidge, D.R.; Chiari, R.; Bearz, A.; Lin, C.-C.; Gadgeel, S.M.; et al. Lorlatinib in Patients with ALK-Positive Non-Small-Cell Lung Cancer: Results from a Global Phase 2 Study. Lancet Oncol. 2018, 19, 1654–1667. [Google Scholar] [CrossRef]
  79. Shaw, A.T.; Bauer, T.M.; de Marinis, F.; Felip, E.; Goto, Y.; Liu, G.; Mazieres, J.; Kim, D.-W.; Mok, T.; Polli, A.; et al. First-Line Lorlatinib or Crizotinib in Advanced ALK-Positive Lung Cancer. N. Engl. J. Med. 2020, 383, 2018–2029. [Google Scholar] [CrossRef]
  80. Skoulidis, F.; Li, B.T.; Dy, G.K.; Price, T.J.; Falchook, G.S.; Wolf, J.; Italiano, A.; Schuler, M.; Borghaei, H.; Barlesi, F.; et al. Sotorasib for Lung Cancers with KRAS p.G12C Mutation. N. Engl. J. Med. 2021, 384, 2371–2381. [Google Scholar] [CrossRef]
  81. Lee, A. Sotorasib: A Review in KRAS G12C Mutation-Positive Non-Small Cell Lung Cancer. Target. Oncol. 2022, 17, 727–733. [Google Scholar] [CrossRef] [PubMed]
  82. Schmid, P.; Cortes, J.; Dent, R.; McArthur, H.; Pusztai, L.; Kümmel, S.; Denkert, C.; Park, Y.H.; Hui, R.; Harbeck, N.; et al. Overall Survival with Pembrolizumab in Early-Stage Triple-Negative Breast Cancer. N. Engl. J. Med. 2024, 391, 1981–1991. [Google Scholar] [CrossRef] [PubMed]
  83. Jänne, P.A.; Riely, G.J.; Gadgeel Shirish, M.; Heist, R.S.; Ou, S.-H.I.; Pacheco, J.M.; Johnson, M.L.; Sabari, J.K.; Leventakos, K.; Yau, E.; et al. Adagrasib in Non-Small-Cell Lung Cancer Harboring a KRASG12C. N. Engl. J. Med. 2022, 387, 120–131. [Google Scholar] [CrossRef] [PubMed]
  84. Ou, S.-H.I.; Jänne, P.A.; Leal Ticiana, A.; Rybkin, I.I.; Sabari, J.K.; Barve, M.A.; Bazhenova, L.; Johnson, M.L.; Velastegui, K.L.; Cilliers, C.; et al. First-in-Human Phase I/IB Dose-Finding Study of Adagrasib(MRTX849) in Patients with Advanced KRASG12C Solid Tumors(KRYSTAL-1). J. Clin. Oncol. 2022, 40, 2530–2538. [Google Scholar] [CrossRef]
  85. Drilon, A.; Oxnard, G.R.; Tan, D.S.W.; Loong, H.H.F.; Johnson, M.; Gainor, J.; McCoach, C.E.; Gautschi, O.; Besse, B.; Cho, B.C.; et al. Efficacy of Selpercatinib in RET Fusion-Positive-Small-Cell Lung Cancer. N. Engl. J. Med. 2020, 383, 813–824. [Google Scholar] [CrossRef]
  86. Drilon, A.; Subbiah, V.; Gautschi, O.; Tomasini, P.; de Braud, F.; Solomon, B.J.; Tan, D.S.-W.; Alonso, G.; Wolf, J.; Park, K.; et al. Selpercatinib in Patients with RET Fusion-Positive Non-Small-Cell Lung Cancer: Updated Safety and Efficacy from the registrational LIBRETTO-001 Phase I/II Trial. J. Clin. Oncol. 2023, 41, 385–394. [Google Scholar] [CrossRef]
  87. Zhou, Q.; Zhao, J.; Chang, J.; Wang, H.; Fan, Y.; Wang, K.; Wu, G.; Nian, W.; Sun, Y.; Sun, M.; et al. Efficacy and Safety of Pralsetinib in Patients with Advanced Fusion-Positive Non-Small Cell Lung Cancer. Cancer 2023, 129, 3239–3251. [Google Scholar] [CrossRef]
  88. Gainor, J.F.; Curigliano, G.; Kim, D.-W.; Lee, D.H.; Besse, B.; Baik, C.S.; Doebele, R.C.; Cassier, P.A.; Lopes, G.; Tan, D.S.W.; et al. Pralsetinib for RET Fusion-Positive Non-Small-Cell Lung Cancer(ARROW): A Multi-Cohort, Open-Label, Phase 1/2 Study. Lancet Oncol. 2021, 22, 959–969. [Google Scholar] [CrossRef]
  89. Vansteenkiste, J.F.; Van De Kerkhove, C.; Wauters, E.; Van Mol, P. Capmatinib for the Treatment of Non-Small Cell Lung Cancer. Expert. Rev. Anticancer. Ther. 2019, 19, 659–671. [Google Scholar] [CrossRef]
  90. Paik, P.K.; Felip, E.; Veillon, R.; Sakai, H.; Cortot, A.B.; Garassino, M.C.; Mazieres, J.; Viteri, S.; Senellart, H.; Van Meerbeeck, J.; et al. Tepotinib in Non-Small-Cell Lung Cancer with MET Exon 14 Mutations. N. Engl. J. Med. 2020, 383, 931–943. [Google Scholar] [CrossRef]
  91. Wu, Y.-L.; Cheng, Y.; Zhou, J.; Lu, S.; Zhang, Y.; Zhao, J.; Kim, D.-W.; Soo, R.A.; Kim, S.-W.; Pan, H.; et al. Tepotinib plus Gefitinib in Patients with EGFR-Mutant-Small-Cell Lung Cancer with MET Overexpression or MET and Acquired Resistance to Previous EGFR (INSIGHT Study): An Open-Label, Phase 1b/2, Multicentre, Randomised Trial. Lancet Respir. Med. 2020, 8, 1132–1143. [Google Scholar] [CrossRef] [PubMed]
  92. Yun, J.; Lee, S.-H.; Kim, S.-Y.; Jeong, S.-Y.; Kim, J.-H.; Pyo, K.-H.; Park, C.-W.; Heo, S.G.; Yun, M.R.; Lim, S.; et al. Antitumor Activity of Amivantamab (JNJ-61186372), an EGFR-MET Bispecific Antibody, in Diverse Models of EGFR Exon Insertion-Driven NSCLC. Cancer Discov. 2020, 10, 1194–1209. [Google Scholar] [CrossRef]
  93. Passaro, A.; Wang, J.; Wang, Y.; Lee, S.-H.; Melosky, B.; Shih, J.-Y.; Wang, J.; Azuma, K.; Juan-Vidal, O.; Cobo, M.; et al. Amivantamab plus Chemotherapy with and without Lazertinib in EGFR-Mutant Advanced NSCLC after Disease Progression on osimertinib: Primary Results from the Phase III MARIPOSA-2 Study. Ann. Oncol. 2024, 35, 77–90. [Google Scholar] [CrossRef] [PubMed]
  94. Riely, G.J.; Neal, J.W.; Camidge, D.R.; Spira, A.I.; Piotrowska, Z.; Costa, D.B.; Tsao, A.S.; Patel, J.D.; Gadgeel, S.M.; Bazhenova, L.; et al. Activity and Safety of Mobocertinib (TAK-788) in Previously Non-Small Cell Lung Cancer with EGFR Exon 20 Insertion from a Phase I/II Trial. Cancer Discov. 2021, 11, 1688–1699. [Google Scholar] [CrossRef]
  95. Zhou, C.; Ramalingam, S.S.; Kim, T.M.; Kim, S.-W.; Yang, J.C.-H.; Riely, G.J.; Mekhail, T.; Nguyen, D.; Garcia Campelo, M.R.; Felip, E.; et al. Treatment Outcomes and Safety of Mobocertinib In-Pretreated Patients with EGFR Exon 20-Positive Metastatic Non-Small Cell Lung Cancer: A 1/2 Open-Label Nonrandomized Clinical Trial. JAMA Oncol. 2021, 7, e214761. [Google Scholar] [CrossRef]
  96. Lee, J.; Park, S.; Jung, H.A.; Sun, J.-M.; Lee, S.-H.; Ahn, J.S.; Park, K.; Ahn, M.-J. Evaluating Entrectinib as a Treatment Option for Non-Small Cell Lung Cancer. Expert. Opin. Pharmacother. 2020, 21, 1935–1942. [Google Scholar] [CrossRef]
  97. Drilon, A.; Siena, S.; Dziadziuszko, R.; Barlesi, F.; Krebs, M.G.; Shaw, A.T.; de Braud, F.; Rolfo, C.; Ahn, M.-J.; Wolf, J.; et al. Entrectinib in ROS1 Fusion-Positive Non-Small-Cell Lung Cancer: Integrated Analysis of Three Phase 1-2 Trials. Lancet Oncol. 2020, 21, 261–270. [Google Scholar] [CrossRef] [PubMed]
  98. Drilon, A.; Tan, D.S.W.; Lassen, U.N.; Leyvraz, S.; Liu, Y.; Patel, J.D.; Rosen, L.; Solomon, B.; Norenberg, R.; Dima, L.; et al. Efficacy and Safety of Larotrectinib in Patients with tropomyosin Receptor Kinase Fusion-Positive Lung Cancers. JCO Precis. Oncol. 2022, 6, e2100418. [Google Scholar] [CrossRef]
  99. Hong, D.S.; Bauer, T.M.; Lee, J.J.; Dowlati, A.; Brose, M.S.; Farago, A.F.; Taylor, M.; Shaw, A.T.; Montez, S.; Meric-Bernstam, F.; et al. Larotrectinib in Adult Patients with Solid Tumours: A-Centre, Open-Label, Phase I Dose-Escalation Study. Ann. Oncol. 2019, 30, 325–331. [Google Scholar] [CrossRef]
  100. Gettinger, S.; Rizvi, N.A.; Chow, L.Q.; Borghaei, H.; Brahmer, J.; Ready, N.; Gerber, D.E.; Shepherd, F.A.; Antonia, S.; Goldman, J.W.; et al. Nivolumab Monotherapy for First-Line Treatment of Advanced Non–Small-Cell Lung Cancer. J. Clin. Oncol. 2016, 34, 2980–2987. [Google Scholar] [CrossRef]
  101. Costantini, A.; Julie, C.; Dumenil, C.; Hélias-Rodzewicz, Z.; Tisserand, J.; Dumoulin, J.; Giraud, V.; Labrune, S.; Chinet, T.; Emile, J.-F.; et al. Predictive Role of Plasmatic Biomarkers in Advanced Non-Small Cell Lung Cancer Treated by Nivolumab. Oncoimmunology 2018, 7, e1452581. [Google Scholar] [CrossRef] [PubMed]
  102. Hellmann, M.D.; Paz-Ares, L.; Bernabe Caro, R.; Zurawski, B.; Kim, S.-W.; Carcereny Costa, E.; Park, K.; Alexandru, A.; Lupinacci, L.; de la Mora Jimenez, E.; et al. Nivolumab plus Ipilimumab in Advanced Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2019, 381, 2020–2031. [Google Scholar] [CrossRef] [PubMed]
  103. 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]
  104. Reck, M. Pembrolizumab as First-Line Therapy for Metastatic Non-Small-Cell Lung Cancer. Immunotherapy 2018, 10, 93–105. [Google Scholar] [CrossRef]
  105. Reck, M.; Rodriguez-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–Small-Cell Lung Cancer. N. Engl. J. Med. 2016, 375, 1823–1833. [Google Scholar] [CrossRef] [PubMed]
  106. Antonia, S.J.; Villegas, A.; Daniel, D.; Vicente, D.; Murakami, S.; Hui, R.; Yokoi, T.; Chiappori, A.; Lee, K.H.; de Wit, M.; et al. Durvalumab after Chemoradiotherapy in Stage III Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2017, 377, 1919–1929. [Google Scholar] [CrossRef]
  107. Rizvi, N.A.; Cho, B.C.; Reinmuth, N.; Lee, K.H.; Luft, A.; Ahn, M.-J.; van den Heuvel, M.M.; Cobo, M.; Vicente, D.; Smolin, A.; et al. Durvalumab with or without Tremelimumab vs Standard chemotherapy in First-Line Treatment of Metastatic Non-Small Cell Lung: The MYSTIC Phase 3 Randomized Clinical Trial. JAMA Oncol. 2020, 6, 661–674. [Google Scholar] [CrossRef]
  108. Sequist, L.V.; Han, J.-Y.; Ahn, M.-J.; Cho, B.C.; Yu, H.; Kim, S.-W.; Yang, J.C.-H.; Lee, J.S.; Su, W.-C.; Kowalski, D.; et al. Osimertinib plus Savolitinib in Patients with EGFR-Positive, MET-Amplified, Non-Small-Cell Lung cancer after Progression on EGFR Tyrosine Kinase Inhibitors: Interim from a Multicentre, Open-Label, Phase 1b Study. Lancet Oncol. 2020, 21, 373–386. [Google Scholar] [CrossRef]
  109. Lu, S.; Fang, J.; Li, X.; Cao, L.; Zhou, J.; Guo, Q.; Liang, Z.; Cheng, Y.; Jiang, L.; Yang, N.; et al. Once-Daily Savolitinib in Chinese Patients with Pulmonary Carcinomas and Other Non-Small-Cell Lung Cancers MET Exon 14 Skipping Alterations: A Multicentre, Single-Arm, Open-Label, Phase 2 Study. Lancet Respir. Med. 2021, 9, 1154–1164. [Google Scholar] [CrossRef]
  110. Ouyang, W.; Hu, J.; Zhang, H.; Xie, C. The Management of Patients with Lung Cancer during the outbreak of Coronavirus Disease 2019. J. Thorac. Oncol. 2020, 15, e106–e107. [Google Scholar] [CrossRef]
  111. Odogwu, L.; Mathieu, L.; Blumenthal, G.; Larkins, E.; Goldberg, K.B.; Griffin, N.; Bijwaard, K.; Lee, E.Y.; Philip, R.; Jiang, X.; et al. FDA Approval Summary: Dabrafenib and Trametinib for the treatment of Metastatic Non-Small Cell Lung Cancers Harboring BRAF V600E Mutations. Oncologist 2018, 23, 740–745. [Google Scholar] [CrossRef] [PubMed]
  112. Planchard, D.; Smit, E.F.; Groen, H.J.M.; Mazieres, J.; Besse, B.; Helland, Å.; Giannone, V.; D’Amelio, A.M., Jr.; Zhang, P.; Mookerjee, B.; et al. Dabrafenib plus Trametinib in Patients with Previously Untreated-Mutant Metastatic Non-Small-Cell Lung Cancer: An-Label, Phase 2 Trial. Lancet Oncol. 2017, 18, 1307–1316. [Google Scholar] [CrossRef] [PubMed]
  113. Cai, L.; Li, Y.; Tan, J.; Xu, L.; Li, Y. Targeting LAG-3, TIM-3, and TIGIT for Cancer Immunotherapy. J. Hematol. Oncol. 2023, 16, 101. [Google Scholar] [CrossRef]
  114. 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]
  115. Al-Batran, S.-E.; Mueller, D.W.; Rafiyan, M.-R.; Kiselicki, D.; Atmaca, A.; Habibzada, T.; Mueller, C.; Brignone, C.; Triebel, F.; Loose, M.; et al. A Soluble LAG-3 Protein (Eftilagimod Alpha) and an anti-PD-L1 Antibody (Avelumab) Tested in a Phase I Trial: A New Combination in Immuno-Oncology. ESMO Open 2023, 8, 101623. [Google Scholar] [CrossRef]
  116. 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-Small Cell Lung Cancer or Melanoma Who Received Prior with Anti-PD-1/PD-L1 Therapy: A Phase 2 Multicentre. BMJ Open 2024, 14, e079132. [Google Scholar] [CrossRef]
  117. Kim, T.W.; Bedard, P.L.; LoRusso, P.; Gordon, M.S.; Bendell, J.; Oh, D.-Y.; Ahn, M.-J.; Garralda, E.; D’Angelo, S.P.; Desai, J.; et al. Anti-TIGIT Antibody Tiragolumab Alone or with Atezolizumab in patients with Advanced Solid Tumors: A Phase 1a/1b Nonrandomized Trial. JAMA Oncol. 2023, 9, 1574–1582. [Google Scholar] [CrossRef] [PubMed]
  118. Naidoo, J.; Peters, S.; Runglodvatana, Y.; Li, J.Y.-C.; Fong, C.H.; Ho, G.F.; How, S.H.; Juengsamarn, J.; Todd, T.; Marina, N.; et al. 1461 Phase 2 Randomized Study of Domvanalimab Combined with zimberelimab in Front-Line, PD-(L)1 High, Locally Advanced or metastatic Non-Small Cell Lung Cancer (NSCLC): Results from ARC-10 Part 1. In Proceedings of the Late-Breaking Abstracts; BMJ Publishing Group Ltd.: London, UK, 2024; pp. A1690–A1691. [Google Scholar]
  119. Kim, C.-G.; Sang, Y.-B.; Lee, J.-H.; Chon, H.-J. Combining Cancer Vaccines with Immunotherapy: Establishing a New Approach. Int. J. Mol. Sci. 2021, 22, 8035. [Google Scholar] [CrossRef]
  120. Le Tourneau, C.; Rischin, D.; Groenland, S.; Lim, A.; Martin-Liberal, J.; Moreno, V.; Trigo, J.M.; Mathew, M.; Cho, D.; Hansen, A.; et al. 1O Inducible T Cell Co-Stimulatory (ICOS) Receptor, GSK3359609 (GSK609) Alone and Combination with pembrolizumab: Preliminary Results from INDUCE-1 Expansion in Head and Neck Squamous Cell Carcinoma (HNSCC). Ann. Oncol. 2020, 31, S1. [Google Scholar] [CrossRef]
  121. Atkins, M.B.; Ascierto, P.A.; Feltquate, D.; Gulley, J.L.; Johnson, D.B.; Khushalani Nikhil, I.; Sosman, J.; Yap, T.A.; Kluger, H.; Sullivan, R.J.; et al. Society for Immunotherapy of Cancer (SITC) Consensus Definitions for Resistance to Combinations of Immune Checkpoint Inhibitors with Targeted Therapies. J. Immunother. Cancer 2023, 11, e005923. [Google Scholar] [CrossRef]
  122. Zhou, S.; Yang, H. Immunotherapy Resistance in Non-Small-Cell Lung Cancer: From Mechanism to Clinical Strategies. Front. Immunol. 2023, 14, 1129465. [Google Scholar] [CrossRef]
  123. West, H.; McCleod, M.; Hussein, M.; Morabito, A.; Rittmeyer, A.; Conter Henry, J.; Kopp, H.-G.; Daniel, D.; McCune, S.; Mekhail, T.; et al. Atezolizumab in Combination with Carboplatin plus Nab-Paclitaxel Compared with Chemotherapy Alone as First-Line for Metastatic Non-Squamous Non-Small-Cell Lung Cancer(IMpower130): A Multicentre, Randomised, Open-Label, Phase 3. Lancet Oncol. 2019, 20, 924–937. [Google Scholar] [CrossRef]
  124. Seto, T.; Sam, D.; Pan, M. Mechanisms of Primary and Secondary Resistance to Immune Checkpoint Inhibitors in Cancer. Med. Sci. 2019, 7, 14. [Google Scholar] [CrossRef] [PubMed]
  125. Said, S.S.; Ibrahim, W.N. Cancer Resistance to Immunotherapy: Comprehensive Insights with future Perspectives. Pharmaceutics 2023, 15, 1143. [Google Scholar] [CrossRef] [PubMed]
  126. Schoenfeld, A.J.; Hellmann, M.D. Acquired Resistance to Immune Checkpoint Inhibitors. Cancer Cell 2020, 37, 443–455. [Google Scholar] [CrossRef] [PubMed]
  127. Abushukair, H.; Ababneh, O.; Zaitoun, S.; Saeed, A. Primary and Secondary Immune Checkpoint Inhibitors Resistance in colorectal Cancer: Key Mechanisms and Ways to Overcome Resistance. Cancer Treat. Res. Commun. 2022, 33, 100643. [Google Scholar] [CrossRef]
  128. Bell, H.N.; Zou, W. Beyond the Barrier: Unraveling the Mechanisms of Immunotherapy Resistance. Annu. Rev. Immunol. 2024, 42, 521–550. [Google Scholar] [CrossRef]
  129. Sharma, P.; Hu-Lieskovan, S.; Wargo Jennifer, A.; Ribas, A. Primary, Adaptive, and Acquired Resistance to Cancer. Cell 2017, 168, 707–723. [Google Scholar] [CrossRef]
  130. Ricciuti, B.; Lamberti, G.; Puchala Sreekar, R.; Mahadevan, N.R.; Lin, J.-R.; Alessi, J.V.; Chowdhury, A.; Li, Y.Y.; Wang, X.; Spurr, L.; et al. Genomic and Immunophenotypic Landscape of Acquired Resistance to PD-(L)1 Blockade in Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2024, 42, 1311–1321. [Google Scholar] [CrossRef]
  131. Ricciuti, B.; Arbour, K.C.; Lin, J.J.; Vajdi, A.; Vokes, N.; Hong, L.; Zhang, J.; Tolstorukov, M.Y.; Li, Y.Y.; Spurr, L.F.; et al. Diminished Efficacy of Programmed Death-(Ligand)1 Inhibition in STK11- and KEAP1-Mutant Lung Adenocarcinoma Is Affected by KRAS Mutation Status. J. Thorac. Oncol. 2022, 17, 399–410. [Google Scholar] [CrossRef]
  132. Qian, Y.; Galan-Cobo, A.; Guijarro, I.; Dang, M.; Molkentine, D.; Poteete, A.; Zhang, F.; Wang, Q.; Wang, J.; Parra, E.; et al. MCT4-Dependent Lactate Secretion Suppresses Antitumor immunity in LKB1-Deficient Lung Adenocarcinoma. Cancer Cell 2023, 41, 1363–1380.e7. [Google Scholar] [CrossRef]
  133. Marinelli, D.; Mazzotta, M.; Scalera, S.; Terrenato, I.; Sperati, F.; D’Ambrosio, L.; Pallocca, M.; Corleone, G.; Krasniqi, E.; Pizzuti, L.; et al. KEAP1-Driven Co-Mutations in Lung Adenocarcinoma unresponsive to Immunotherapy despite High Tumor Mutational Burden. Ann. Oncol. 2020, 31, 1746–1754. [Google Scholar] [CrossRef]
  134. Tanaka, I.; Koyama, J.; Itoigawa, H.; Hayai, S.; Morise, M. Metabolic Barriers in Non-Small Cell Lung Cancer with LKB1 and/or KEAP1 Mutations for Immunotherapeutic Strategies. Front. Oncol. 2023, 13, 1249237. [Google Scholar] [CrossRef]
  135. Best, S.A.; Gubser, P.M.; Sethumadhavan, S.; Kersbergen, A.; Negrón Abril, Y.L.; Goldford, J.; Sellers, K.; Abeysekera, W.; Garnham, A.L.; McDonald, J.A.; et al. Glutaminase Inhibition Impairs CD8 T Cell Activation in STK11-/Lkb1-Deficient Lung Cancer. Cell Metab. 2022, 34, 874–887.e6. [Google Scholar] [CrossRef]
  136. Zavitsanou, A.-M.; Pillai, R.; Hao, Y.; Wu, W.L.; Bartnicki, E.; Karakousi, T.; Rajalingam, S.; Herrera, A.; Karatza, A.; Rashidfarrokhi, A.; et al. KEAP1 Mutation in Lung Adenocarcinoma Promotes Immune evasion and Immunotherapy Resistance. Cell Rep. 2023, 42, 113295. [Google Scholar] [CrossRef]
  137. Fountzilas, E.; Kurzrock, R.; Vo, H.H.; Tsimberidou, A.-M. Wedding of Molecular Alterations and Immune Checkpoint Blockade: Genomics as a Matchmaker. J. Natl. Cancer Inst. 2021, 113, 1634–1647. [Google Scholar] [CrossRef]
  138. Ramapriyan, R.; Caetano, M.S.; Barsoumian, H.B.; Mafra, A.C.P.; Zambalde, E.P.; Menon, H.; Tsouko, E.; Welsh James, W.; Cortez, M.A. Altered Cancer Metabolism in Mechanisms of Immunotherapy Resistance. Pharmacol. Ther. 2019, 195, 162–171. [Google Scholar] [CrossRef]
  139. Otegui, N.; Houry, M.; Arozarena, I.; Serrano, D.; Redin, E.; Exposito, F.; Leon, S.; Valencia, K.; Montuenga, L.; Calvo, A. Cancer Cell-Intrinsic Alterations Associated with an immunosuppressive Tumor Microenvironment and Resistance to immunotherapy in Lung Cancer. Cancers 2023, 15, 3076. [Google Scholar] [CrossRef]
  140. Koyama, S.; Akbay, E.A.; Li, Y.Y.; Aref, A.R.; Skoulidis, F.; Herter-Sprie, G.S.; Buczkowski, K.A.; Liu, Y.; Awad, M.M.; Denning, W.L.; et al. STK11/LKB1 Deficiency Promotes Neutrophil Recruitment and proinflammatory Cytokine Production to Suppress T-Cell activity in the Lung Tumor Microenvironment. Cancer Res. 2016, 76, 999–1008. [Google Scholar] [CrossRef]
  141. Iwan, E.; Grenda, A.; Bomba, A.; Bielińska, K.; Wasyl, D.; Kieszko, R.; Rolska-Kopińska, A.; Chmielewska, I.; Krawczyk, P.; Rybczyńska-Tkaczyk, K.; et al. Gut Resistome of NSCLC Patients Treated with Immunotherapy. Front. Genet. 2024, 15, 1378900. [Google Scholar] [CrossRef]
  142. Xiao, Y.; Yu, D. Tumor Microenvironment as a Therapeutic Target in Cancer. Pharmacol. Ther. 2021, 221, 107753. [Google Scholar] [CrossRef]
  143. Wong, K.Y.; Cheung, A.H.-K.; Chen, B.; Chan, W.N.; Yu, J.; Lo, K.W.; Kang, W.; To, K.F. Cancer-Associated Fibroblasts in Nonsmall Cell Lung Cancer: From Molecular Mechanisms to Clinical Implications. Int. J. Cancer 2022, 151, 1195–1215. [Google Scholar] [CrossRef] [PubMed]
  144. Guo, T.; Xu, J. Cancer-Associated Fibroblasts: A Versatile Mediator in Tumor Progression, Metastasis, and Targeted Therapy. Cancer Metastasis Rev. 2024, 43, 1095–1116. [Google Scholar] [CrossRef] [PubMed]
  145. Hinshaw, D.C.; Shevde, L.A. The Tumor Microenvironment Innately Modulates Cancer Progression. Cancer Res. 2019, 79, 4557–4566. [Google Scholar] [CrossRef] [PubMed]
  146. Huang, S.; Chung, J.Y.-F.; Li, C.; Wu, Y.; Qiao, G.; To, K.-F.; Tang, P.M.-K. Cellular Dynamics of Tumor Microenvironment Driving Immunotherapy Resistance in Non-Small-Cell Lung Carcinoma. Cancer Lett. 2024, 604, 217272. [Google Scholar] [CrossRef]
  147. Miao, Y.; Wang, W.; Dong, Y.; Hu, J.; Wei, K.; Yang, S.; Lai, X.; Tang, H. Hypoxia Induces Tumor Cell Growth and Angiogenesis in Non-Small Cell Lung Carcinoma via the Akt-PDK1-HIF1α-YKL-40 Pathway. Transl. Cancer Res. 2020, 9, 2904–2918. [Google Scholar] [CrossRef]
  148. Ngaha, T.Y.S.; Zhilenkova, A.V.; Essogmo, F.E.; Uchendu, I.K.; Abah, M.O.; Fossa, L.T.; Sangadzhieva, Z.D.; D Sanikovich, V.; S Rusanov, A.; N Pirogova, Y.; et al. Angiogenesis in Lung Cancer: Understanding the Roles of Growth Factors. Cancers 2023, 15, 4648. [Google Scholar] [CrossRef]
  149. Bielenberg, D.R.; Zetter, B.R. The Contribution of Angiogenesis to the Process of Metastasis. Cancer J. 2015, 21, 267–273. [Google Scholar] [CrossRef]
  150. Chen, G.; Wu, K.; Li, H.; Xia, D.; He, T. Role of Hypoxia in the Tumor Microenvironment and Targeted Therapy. Front. Oncol. 2022, 12, 961637. [Google Scholar] [CrossRef]
  151. Parker, A.L.; Cox, T.R. The Role of the ECM in Lung Cancer Dormancy and Outgrowth. Front. Oncol. 2020, 10, 1766. [Google Scholar] [CrossRef]
  152. Saito, A.; Horie, M.; Nagase, T. TGF-β Signaling in Lung Health and Disease. Int. J. Mol. Sci. 2018, 19, 2460. [Google Scholar] [CrossRef] [PubMed]
  153. Li, F.; Zheng, L.; Xu, X.; Jin, J.; Li, X.; Zhou, L. The Impact of Chronic Obstructive Pulmonary Disease on the Risk of Immune-Related Pneumonitis in Lung Cancer Patients Undergoing Immunotherapy: A Systematic Review and Meta-Analysis. BMC Pulm. Med. 2024, 24, 393. [Google Scholar] [CrossRef] [PubMed]
  154. Zhou, P.; Zhao, X.; Wang, G. Risk Factors for Immune Checkpoint Inhibitor-Related Pneumonitis in Cancer Patients: A Systemic Review and Meta-Analysis. Respiration 2022, 101, 1035–1050. [Google Scholar] [CrossRef]
  155. Mark, N.M.; Kargl, J.; Busch, S.E.; Yang, G.H.Y.; Metz, H.E.; Zhang, H.; Hubbard, J.J.; Pipavath, S.N.J.; Madtes David, K.; Houghton, A.M. Chronic Obstructive Pulmonary Disease Alters Immune Cell Composition and Immune Checkpoint Inhibitor Efficacy In-Small Cell Lung Cancer. Am. J. Respir. Crit. Care Med. 2018, 197, 325–336. [Google Scholar] [CrossRef]
  156. Zeng, X.; Zhu, S.; Xu, C.; Wang, Z.; Su, X.; Zeng, D.; Long, H.; Zhu, B. Effect of Comorbidity on Outcomes of Patients with Advanced Non-Small Cell Lung Cancer Undergoing Anti-PD1 Immunotherapy. Med. Sci. Monit. 2020, 26, e922576. [Google Scholar] [CrossRef]
  157. Jaiyesimi, I.A.; Leighl, N.B.; Ismaila, N.; Alluri, K.; Florez, N.; Gadgeel, S.; Masters, G.; Schenk, E.L.; Schneider, B.J.; Sequist, L.; et al. Therapy for Stage IV Non-Small Cell Lung Cancer without Driver: ASCO Living Guideline, Version 2023.3. J. Clin. Oncol. 2024, 42, e23–e43. [Google Scholar] [CrossRef]
  158. Hamilton, G.; Rath, B. Immunotherapy for Small Cell Lung Cancer: Mechanisms Of. Expert. Opin. Biol. Ther. 2019, 19, 423–432. [Google Scholar] [CrossRef]
  159. Shah, S.; Wood, K.; Labadie, B.; Won, B.; Brisson, R.; Karrison, T.; Hensing, T.; Kozloff, M.; Bao, R.; Patel, J.D.; et al. Clinical and Molecular Features of Innate and Acquired to Anti-PD-1/PD-L1 Therapy in Lung Cancer. Oncotarget 2018, 9, 4375–4384. [Google Scholar] [CrossRef] [PubMed]
  160. Isomoto, K.; Haratani, K.; Tsujikawa, T.; Makutani, Y.; Kawakami, H.; Takeda, M.; Yonesaka, K.; Tanaka, K.; Iwasa, T.; Hayashi, H.; et al. Mechanisms of Primary and Acquired Resistance to Immune Inhibitors in Advanced Non-Small Cell Lung Cancer: A Immunohistochemistry-Based Single-Cell Analysis. Lung Cancer 2022, 174, 71–82. [Google Scholar] [CrossRef]
  161. Hiltbrunner, S.; Cords, L.; Kasser, S.; Freiberger, S.N.; Kreutzer, S.; Toussaint Nora, C.; Grob, L.; Opitz, I.; Messerli, M.; Zoche, M.; et al. Acquired Resistance to Anti-PD1 Therapy in Patients with NSCLC Associates with Immunosuppressive T Cell Phenotype. Nat. Commun. 2023, 14, 5154. [Google Scholar] [CrossRef]
  162. Kim, Y.; Kim, C.H.; Lee, H.Y.; Lee, S.-H.; Kim, H.S.; Lee, S.; Cha, H.; Hong, S.; Kim, K.; Seo, S.W.; et al. Comprehensive Clinical and Genetic Characterization of hyperprogression Based on Volumetry in Advanced Non-Small Cell Cancer Treated with Immune Checkpoint Inhibitor. J. Thorac. Oncol. 2019, 14, 1608–1618. [Google Scholar] [CrossRef]
  163. Li, G.; Choi, J.E.; Kryczek, I.; Sun, Y.; Liao, P.; Li, S.; Wei, S.; Grove, S.; Vatan, L.; Nelson, R.; et al. Intersection of Immune and Oncometabolic Pathways Drives Cancer during Immunotherapy. Cancer Cell 2023, 41, 304–322.e7. [Google Scholar] [CrossRef] [PubMed]
  164. Salous, T.; Shukla, N.A.; Althouse, S.K.; Perkins, S.M.; Furqan, M.; Leal, T.; Traynor, A.M.; Feldman, L.E.; Hanna, N.H.; Durm, G.A. A Phase 2 Trial of Chemotherapy plus Pembrolizumab in patients with Advanced Non-Small Cell Lung Cancer Previously Treated with a PD-1 or PD-L1 Inhibitor: Big Ten Cancer Research BTCRC-LUN15-029. Cancer 2023, 129, 264–271. [Google Scholar] [CrossRef] [PubMed]
  165. Meyer, M.-L.; Fitzgerald, B.G.; Paz-Ares, L.; Cappuzzo, F.; Jänne, P.A.; Peters, S.; Hirsch, F.R. New Promises and Challenges in the Treatment of Advanced Non-Small-Cell Lung Cancer. Lancet 2024, 404, 803–822. [Google Scholar] [CrossRef]
  166. Wang, Y.-J.; Fletcher, R.; Yu, J.; Zhang, L. Immunogenic Effects of Chemotherapy-Induced Tumor Cell Death. Genes Dis. 2018, 5, 194–203. [Google Scholar] [CrossRef] [PubMed]
  167. Yu, S.; Wang, Y.; He, P.; Shao, B.; Liu, F.; Xiang, Z.; Yang, T.; Zeng, Y.; He, T.; Ma, J.; et al. Effective Combinations of Immunotherapy and Radiotherapy for cancer Treatment. Front. Oncol. 2022, 12, 809304. [Google Scholar] [CrossRef]
  168. Spigel, D.; Jotte, R.; Nemunaitis, J.; Shum, M.; Schneider, J.; Goldschmidt, J.; Eisenstein, J.; Berz, D.; Seneviratne, L.; Socoteanu, M.; et al. Randomized Phase 2 Studies of Checkpoint Inhibitors Alone or in combination with Pegilodecakin in Patients with Metastatic NSCLC (CYPRESS 1 and CYPRESS 2). J. Thorac. Oncol. 2021, 16, 327–333. [Google Scholar] [CrossRef]
  169. Zhang, X.; Wang, X.; Hou, L.; Xu, Z.; Liu, Y.; Wang, X. Nanoparticles Overcome Adaptive Immune Resistance and Enhance Immunotherapy via Targeting Tumor Microenvironment in Lung Cancer. Front. Pharmacol. 2023, 14, 1130937. [Google Scholar] [CrossRef]
  170. Herbst, R.S.; Majem, M.; Barlesi, F.; Carcereny, E.; Chu, Q.; Monnet, I.; Sanchez-Hernandez, A.; Dakhil, S.; Camidge, D.R.; Winzer, L.; et al. COAST: An Open-Label, Phase II, Multidrug Platform Study of durvalumab Alone or in Combination with Oleclumab or monalizumab in Patients with Unresectable, Stage III Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2022, 40, 3383–3393. [Google Scholar] [CrossRef]
  171. Yuan, J.; Khilnani, A.; Brody, J.; Andtbacka, R.H.I.; Hu-Lieskovan, S.; Luke Jason, J.; Diab, A.; Marabelle, A.; Snyder, A.; Cao, Z.A.; et al. Current Strategies for Intratumoural Immunotherapy—Beyond immune Checkpoint Inhibition. Eur. J. Cancer 2021, 157, 493–510. [Google Scholar] [CrossRef]
  172. Kim, J.; Hong, J.; Lee, J.; Fakhraei Lahiji, S.; Kim, Y.-H. Recent Advances in Tumor Microenvironment-Targeted Nanomedicine Delivery Approaches to Overcome Limitations of Immune Checkpoint-Based Immunotherapy. J. Control. Release 2021, 332, 109–126. [Google Scholar] [CrossRef] [PubMed]
  173. Isla, D.; Sánchez, A.; Casal, J.; Cobo, M.; Majem, M.; Reguart, N.; Zugazagoitia, J.; Bernabé, R. PD-1/PD-L1 Inhibitors as Monotherapy in the First-Line Treatment of Advanced Non-Small Cell Lung Cancer Patients with high PD-L1 Expression: An Expert Position Statement. J. Clin. Med. 2023, 12, 5063. [Google Scholar] [CrossRef]
  174. Zhou, G.-W.; Xiong, Y.; Chen, S.; Xia, F.; Li, Q.; Hu, J. Anti-PD-1/PD-L1 Antibody Therapy for Pretreated Advanced Nonsmall-Cell Lung Cancer. Medicine 2016, 95, e4611. [Google Scholar] [CrossRef]
  175. Wang, H.; Zhang, Y.; Zhang, H.; Cao, H.; Mao, J.; Chen, X.; Wang, L.; Zhang, N.; Luo, P.; Xue, J.; et al. Liquid Biopsy for Human Cancer: Cancer Screening, Monitoring, and Treatment. MedComm 2024, 5, e564. [Google Scholar] [CrossRef] [PubMed]
  176. Pandey, S.; Yadav, P. Liquid Biopsy in Cancer Management: Integrating Diagnostics and Clinical Applications. Pract. Lab. Med. 2025, 43, e00446. [Google Scholar] [CrossRef]
  177. Fatima, S.; Ma, Y.; Safrachi, A.; Haider, S.; Spring, K.J.; Vafaee, F.; Scott, K.F.; Roberts, T.L.; Becker, T.M.; de Souza, P. Harnessing Liquid Biopsies to Guide Immune Checkpoint Inhibitor Therapy. Cancers 2022, 14, 1669. [Google Scholar] [CrossRef] [PubMed]
  178. Adhit, K.K.; Wanjari, A.; Menon, S.; K, S. Liquid Biopsy: An Evolving Paradigm for Non-Invasive Disease Diagnosis and Monitoring in Medicine. Cureus 2023, 15, e50176. [Google Scholar] [CrossRef]
  179. Borea, R.; Reduzzi, C. The Growing Field of Liquid Biopsy and Its Snowball Effect on Reshaping Cancer Management. J. Liq. Biopsy 2025, 8, 100293. [Google Scholar] [CrossRef]
  180. Bao, Y.; Zhang, D.; Guo, H.; Ma, W. Beyond Blood: Advancing the Frontiers of Liquid Biopsy in Oncology and Personalized Medicine. Cancer Sci. 2024, 115, 1060–1072. [Google Scholar] [CrossRef]
  181. Rother, C.; John, T.; Wong, A. Biomarkers for Immunotherapy Resistance in Non-Small Cell Lung Cancer. Front. Oncol. 2024, 14, 1489977. [Google Scholar] [CrossRef]
  182. Frisone, D.; Friedlaender, A.; Addeo, A.; Tsantoulis, P. The Landscape of Immunotherapy Resistance in NSCLC. Front. Oncol. 2022, 12, 817548. [Google Scholar] [CrossRef]
  183. Agarwala, Y.; Brauns, T.A.; Sluder, A.E.; Poznansky, M.C.; Gemechu, Y. Targeting Metabolic Pathways to Counter Cancer Immunotherapy Resistance. Trends Immunol. 2024, 45, 486–494. [Google Scholar] [CrossRef] [PubMed]
  184. Korn, E.L.; Freidlin, B. Adaptive Clinical Trials: Advantages and Disadvantages of various Adaptive Design Elements. J. Natl. Cancer Inst. 2017, 109, djx013. [Google Scholar] [CrossRef]
  185. Ghorani, E.; Quartagno, M.; Blackhall, F.; Gilbert, D.C.; O’Brien, M.; Ottensmeier, C.; Pizzo, E.; Spicer, J.; Williams, A.; Badman, P.; et al. REFINE-Lung Implements a Novel Multi-Arm Randomised Trial Design to Address Possible Immunotherapy Overtreatment. Lancet Oncol. 2023, 24, e219–e227. [Google Scholar] [CrossRef] [PubMed]
  186. Siu, L.L.; Ivy, S.P.; Dixon, E.L.; Gravell, A.E.; Reeves, S.A.; Rosner, G.L. Challenges and Opportunities in Adapting Clinical Trial design for Immunotherapies. Clin. Cancer Res. 2017, 23, 4950–4958. [Google Scholar] [CrossRef]
  187. Boyer, M.; Şendur, M.A.N.; Rodriguez-Abreu, D.; Park, K.; Lee, D.H.; Çiçin, I.; Yumuk, P.F.; Orlandi, F.J.; Leal, T.A.; Molinier, O.; et al. Pembrolizumab plus Ipilimumab or Placebo for Metastatic-Small-Cell Lung Cancer with PD-L1 Tumor Proportion Score 50%: Randomized, Double-Blind Phase III KEYNOTE-598. J. Clin. Oncol. 2021, 39, 2327–2338. [Google Scholar] [CrossRef] [PubMed]
  188. Senan, S.; Okamoto, I.; Lee, G.-W.; Chen, Y.; Niho, S.; Mak, G.; Yao, W.; Shire, N.; Jiang, H.; Cho, B.C. Design and Rationale for a Phase III, Randomized, Placebo-Controlled Trial of Durvalumab with or without tremelimumab after Concurrent Chemoradiotherapy for patients with Limited-Stage Small-Cell Lung Cancer: The ADRIATIC Study. Clin. Lung Cancer 2020, 21, e84–e88. [Google Scholar] [CrossRef] [PubMed]
  189. Liu, S.V.; Reck, M.; Mansfield, A.S.; Mok, T.; Scherpereel, A.; Reinmuth, N.; Garassino, M.C.; De Castro Carpeno, J.; Califano, R.; Nishio, M.; et al. Updated Overall Survival and PD-L1 Subgroup Analysis of patients with Extensive-Stage Small-Cell Lung Cancer treated with Atezolizumab, Carboplatin, and Etoposide (IMpower133). J. Clin. Oncol. 2021, 39, 619–630. [Google Scholar] [CrossRef]
  190. Peters, S.; Pujol, J.-L.; Dafni, U.; Dómine, M.; Popat, S.; Reck, M.; Andrade, J.; Becker, A.; Moro-Sibilot, D.; Curioni-Fontecedro, A.; et al. Consolidation Nivolumab and Ipilimumab versus Observation in Limited-Disease Small-Cell Lung Cancer after Chemo-Radiotherapy- Results from the Randomised Phase II ETOP/IFCT 4-12 STIMULI Trial. Ann. Oncol. 2022, 33, 67–79. [Google Scholar] [CrossRef]
  191. Neal, J.; Pavlakis, N.; Kim, S.-W.; Goto, Y.; Lim, S.M.; Mountzios, G.; Fountzilas, E.; Mochalova, A.; Christoph, D.C.; Bearz, A.; et al. CONTACT-01: A Randomized Phase III Trial of Atezolizumab + Cabozantinib versus Docetaxel for Metastatic Non-Small Cell Lung Cancer after a Checkpoint Inhibitor and Chemotherapy. J. Clin. Oncol. 2024, 42, 2393–2403. [Google Scholar] [CrossRef]
  192. Borghaei, H.; de Marinis, F.; Dumoulin, D.; Reynolds, C.; Theelen, W.S.M.E.; Percent, I.; Gutierrez Calderon, V.; Johnson, M.L.; Madroszyk-Flandin, A.; Garon, E.B.; et al. SAPPHIRE: Phase III Study of Sitravatinib plus Nivolumab versus Docetaxel in Advanced Nonsquamous Non-Small-Cell Lung Cancer. Ann. Oncol. 2024, 35, 66–76. [Google Scholar] [CrossRef] [PubMed]
  193. George, J.; Maas, L.; Abedpour, N.; Cartolano, M.; Kaiser, L.; Fischer, R.N.; Scheel, A.H.; Weber, J.-P.; Hellmich, M.; Bosco, G.; et al. Evolutionary Trajectories of Small Cell Lung Cancer under Therapy. Nature 2024, 627, 880–889. [Google Scholar] [CrossRef] [PubMed]
  194. Cheng, N.; Liu, J.; Chen, C.; Zheng, T.; Li, C.; Huang, J. Prediction of Lung Cancer Metastasis by Gene Expression. Comput. Biol. Med. 2023, 153, 106490. [Google Scholar] [CrossRef] [PubMed]
  195. Altorki, N.K.; Markowitz, G.J.; Gao, D.; Port, J.L.; Saxena, A.; Stiles, B.; McGraw, T.; Mittal, V. The Lung Microenvironment: An Important Regulator of Tumour Growth and Metastasis. Nat. Rev. Cancer 2019, 19, 9–31. [Google Scholar] [CrossRef]
  196. Wang, L.; Jia, Q.; Chu, Q.; Zhu, B. Targeting Tumor Microenvironment for Non-Small Cell Lung Cancer Immunotherapy. Chin. Med. J. Pulm. Crit. Care Med. 2023, 1, 18–29. [Google Scholar] [CrossRef]
Figure 1. Typology of lung cancer currently used in the clinic, based on histological features.
Figure 1. Typology of lung cancer currently used in the clinic, based on histological features.
Cells 14 00587 g001
Figure 2. Microenvironment of the lung tumour, depicting the level of cancer heterogeneity, complicated intricacies between different cellular and noncellular components, intratumoural hypoxia regions, as well as the possibility of further metastasis due to the free tumour circulating cells crossing the basement membrane into the bloodstream.
Figure 2. Microenvironment of the lung tumour, depicting the level of cancer heterogeneity, complicated intricacies between different cellular and noncellular components, intratumoural hypoxia regions, as well as the possibility of further metastasis due to the free tumour circulating cells crossing the basement membrane into the bloodstream.
Cells 14 00587 g002
Table 3. Results of Checkpoint Inhibitor Trials Targeting LAG-3, TIM-3, and Other Emerging Pathways.
Table 3. Results of Checkpoint Inhibitor Trials Targeting LAG-3, TIM-3, and Other Emerging Pathways.
TargetDrugTrial PhasePopulationKey Findings
LAG-3RelatlimabPhase I/IIAdvanced NSCLCCombined with nivolumab: improved progression-free survival (PFS) in PD-L1-positive tumours. Median PFS: 6.4 months.
LAG-3Eftilagimod alpha (IMP321)Phase IINSCLC, first-lineCombined with pembrolizumab: 47% disease control rate (DCR), well-tolerated.
TIM-3Sabatolimab (MBG453)Phase IAdvanced solid tumours, including NSCLCEarly safety and efficacy data suggest manageable toxicity; limited antitumour activity as monotherapy.
TIM-3TSR-022Phase I/IINSCLCCombined with anti-PD-1: objective response rate (ORR) 25% in heavily pretreated patients.
TIGITTiragolumabPhase IINSCLC, PD-L1-positiveCombined with atezolizumab: PFS benefit (5.6 months vs. 3.9 months) compared to atezolizumab alone.
TIGITDomvanalimabPhase IIAdvanced NSCLCCombined with zimberelimab: ORR of 27.3%, encouraging safety profile.
ICOSGSK3359609Phase IAdvanced solid tumours, including NSCLCPartial responses observed; limited monotherapy efficacy but potential in combinations.
Table 4. Metabolic changes and therapeutic strategies of STK11 (LKB1), KEAP1, and JAK1/2 Mutations.
Table 4. Metabolic changes and therapeutic strategies of STK11 (LKB1), KEAP1, and JAK1/2 Mutations.
MutationMetabolic ChangesTherapeutic Strategies
STK11
(LKB1)
  • epigenetic changes (methylation, histone acetylation)
  • increased lactate production
  • M2 macrophage polarization and T cell dysfunction
  • DNA methyltransferase inhibitors (DNMTis) or histone deacetylase inhibitors (HDACis)
KEAP1
  • hyperactivation of the NRF2 pathway
  • increasing the consumption of glucose and glutamine by tumour cells
  • T cell deprived of nutrients
  • combining ICIs with glutaminase inhibitors
  • epigenetic regulation of the NRF2 pathway
JAK1/2
  • disrupt interferon-gamma signalling
  • agents that enhance antigen presentation or T cell activation
Table 5. Summary of recent clinical trials targeting resistance to immunotherapy in lung cancer.
Table 5. Summary of recent clinical trials targeting resistance to immunotherapy in lung cancer.
Trial Name and ReferenceCancer TypeInvestigated Treatment RegimenPhaseStudy PopulationResultsOvercoming Resistance Strategy
CheckMate 227
[157]
NSCLCNivolumab + Ipilimumab vs. ChemotherapyIIIPatients with Stage IV or recurrent NSCLC who had not received prior systemic therapySignificantly improved overall survival on combination of nivolumab and ipilimumab compared to chemotherapy aloneMultiple immune checkpoints
KEYNOTE-598
[187]
NSCLCPembrolizumab + Ipilimumab vs. PembrolizumabIIIPatients with previously untreated metastatic NSCLC with a PD-L1 tumour proportion score (TPS) ≥ 50% and no sensitising EGFR or ALK aberrationsLack of improved efficacy and greater toxicity of combination therapyMultiple immune checkpoints
ADRIATIC [188]SCLCICI maintenance therapy—Durvalumab (+Tremelimumab)IIIPatients with limited-stage small-cell lung cancer (LS-SCLC) who have not progressed after concurrent chemoradiotherapy (cCRT)Trial is ongoingMultiple immune checkpoints
IMpower133
[189]
SCLCAtezolizumab + Carboplatin + Etoposide vs. Carboplatin + EtoposideIIIPatients with previously untreated extensive-stage small-cell (ES-SCLC) lung cancerCombination of atezolizumab with carboplatin and etoposide as a new standard of care for first-line treatment of ES-SCLC.Enhanced immune response
STIMULI
[190]
SCLCConsolidation Immunotherapy with Nivolumab + IpilimumabIIPatients with limited-stage small-cell lung cancer (LS-SCLC) who had not progressed after concurrent chemoradiotherapy (cCRT) and prophylactic cranial irradiation (PCI)The trial did not meet its primary endpoint of improving progression-free survival (PFS)Multiple immune checkpoints
CONTACT-01
[191]
NSCLCAtezolizumab + Cabozantinib vs. DocetaxelIIIPatients with metastatic non-small cell lung cancer (NSCLC) who had progressed after prior treatment with a checkpoint inhibitor and platinum-containing chemotherapyNot significantly improved OS but significantly improved PFSCombining immune checkpoint inhibition with the anti-angiogenic and immunomodulatory effects of cabozatinib
SAPPHIRE
[192]
NSCLCSitravatinib + Nivolumab vs. DocetaxelIIIPatients with advanced nonsquamous non-small cell lung cancer (NSCLC) who had progressed after prior treatment with a checkpoint inhibitor and platinum-containing chemotherapyNo significant improvement of OS or PFS in Sitravatinib + Nivolumab group compared to DocetaxelCombining immune checkpoint inhibition with the with immunomodulatory effects of sitravatinib
Table 6. Key points regarding genomic medicine in lung cancer—examples from the current landscape and future directions [52,67,70,77,110].
Table 6. Key points regarding genomic medicine in lung cancer—examples from the current landscape and future directions [52,67,70,77,110].
EGFR mutationsPatients with activating EGFR mutations (e.g., exon 19 deletion, L858R) respond well to EGFR TKIs like Gefitinib and Erlotinib. Mutations like T790M drive resistance, but Osimertinib is effective against this mutation.
ALK rearrangementsFound in about 3–7% of NSCLC, these rearrangements respond to ALK inhibitors like Alectinib and Crizotinib. Newer agents like Lorlatinib work against ALK inhibitor-resistant mutations.
KRAS mutationsThe KRAS G12C mutation was historically undruggable, but Sotorasib and Adagrasib are now effective therapies for this subset.
MET exon 14 skippingTherapies like Capmatinib and Tepotinib target this mutation, which occurs in about 3–4% of NSCLC.
RET and ROS1 fusionsDrugs such as Selpercatinib and Crizotinib target fusions in these genes. Entrectinib also targets NTRK gene fusions, which occur less frequently.
PD-1/PD-L1 inhibitorsImmunotherapies like Pembrolizumab, Nivolumab, and Atezolizumab are approved based on high PD-L1 expression and have become standard treatments for NSCLC.
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

Zieliński, P.; Stępień, M.; Chowaniec, H.; Kalyta, K.; Czerniak, J.; Borowczyk, M.; Dwojak, E.; Mroczek, M.; Dworacki, G.; Ślubowska, A.; et al. Resistance in Lung Cancer Immunotherapy and How to Overcome It: Insights from the Genetics Perspective and Combination Therapies Approach. Cells 2025, 14, 587. https://doi.org/10.3390/cells14080587

AMA Style

Zieliński P, Stępień M, Chowaniec H, Kalyta K, Czerniak J, Borowczyk M, Dwojak E, Mroczek M, Dworacki G, Ślubowska A, et al. Resistance in Lung Cancer Immunotherapy and How to Overcome It: Insights from the Genetics Perspective and Combination Therapies Approach. Cells. 2025; 14(8):587. https://doi.org/10.3390/cells14080587

Chicago/Turabian Style

Zieliński, Paweł, Maria Stępień, Hanna Chowaniec, Kateryna Kalyta, Joanna Czerniak, Martyna Borowczyk, Ewa Dwojak, Magdalena Mroczek, Grzegorz Dworacki, Antonina Ślubowska, and et al. 2025. "Resistance in Lung Cancer Immunotherapy and How to Overcome It: Insights from the Genetics Perspective and Combination Therapies Approach" Cells 14, no. 8: 587. https://doi.org/10.3390/cells14080587

APA Style

Zieliński, P., Stępień, M., Chowaniec, H., Kalyta, K., Czerniak, J., Borowczyk, M., Dwojak, E., Mroczek, M., Dworacki, G., Ślubowska, A., Markiewicz, H., Ałtyn, R., & Dobosz, P. (2025). Resistance in Lung Cancer Immunotherapy and How to Overcome It: Insights from the Genetics Perspective and Combination Therapies Approach. Cells, 14(8), 587. https://doi.org/10.3390/cells14080587

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