Next Article in Journal
Prophylactic Treatment of Hepatitis C Virus Infection After Kidney Transplantation with the Combination of Glecaprevir/Pibrentasvir and Sofosbuvir in a Highly Sensitized Hepatitis C Virus-Negative Recipient: A Case Report and Review of the Literature
Previous Article in Journal
Suspected Mitochondrial Dysfunction and Complex Pathophysiology in Fatal Hypermobile Ehlers–Danlos Syndrome: Insights from a Case Report and Post-Mortem Findings
Previous Article in Special Issue
The Tight Relationship Between the Tumoral Microenvironment and Radium-223
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting the Tumor Microenvironment in EGFR-Mutant Lung Cancer: Opportunities and Challenges

1
Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
2
Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
3
Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
4
Division of Pulmonology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
5
Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Institute of Chest Diseases, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
6
Division of Pulmonary Medicine and Allergy, Department of Internal Medicine, Hanyang University College of Medicine, Seoul 04763, Republic of Korea
7
Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea
8
Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 44610, Republic of Korea
9
Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 03083, Republic of Korea
10
Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
11
Department of Precision Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
*
Author to whom correspondence should be addressed.
Biomedicines 2025, 13(2), 470; https://doi.org/10.3390/biomedicines13020470
Submission received: 10 January 2025 / Revised: 11 February 2025 / Accepted: 13 February 2025 / Published: 14 February 2025
(This article belongs to the Special Issue Feature Reviews in Precision Oncology)

Abstract

:
Tyrosine kinase inhibitors (TKIs) have transformed the treatment of epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer. However, treatment resistance remains a major challenge in clinical practice. The tumor microenvironment (TME) is a complex system composed of tumor cells, immune and non-immune cells, and non-cellular components. Evidence indicates that dynamic changes in TME during TKI treatment are associated with the development of resistance. Research has focused on identifying how each component of the TME interacts with tumors and TKIs to understand therapeutic targets that could address TKI resistance. In this review, we describe how TME components, such as immune cells, fibroblasts, blood vessels, immune checkpoint proteins, and cytokines, interact with EGFR-mutant tumors and how they can promote resistance to TKIs. Furthermore, we discuss potential strategies targeting TME as a novel therapeutic approach.

1. Introduction

Lung cancer is the most fatal form of cancer worldwide, with 2.2 million new diagnoses and 1.79 million deaths annually. It is primarily categorized into two histological types: non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC), with NSCLC accounting for 85% of the cases [1]. NSCLC, comprising adenocarcinoma, squamous cell carcinoma, and large cell carcinoma, presents diverse molecular profiles and treatment responses [2]. Recent advancements in targeted therapies have substantially improved the outcomes of patients with advanced NSCLC harboring driver genetic alterations. Epidermal growth factor receptor (EGFR) is the most common targetable driver gene, with mutation incidence of up to 60% in Asian NSCLC populations, which is significantly higher than that in Western populations (10–15%) [3,4]. Targeted therapies have significantly advanced the treatment of NSCLC by selectively inhibiting oncogenic driver mutations, leading to improved patient outcomes (Table 1) [5]. Various clinical trials and real-world studies have demonstrated the survival benefits of EGFR tyrosine kinase inhibitors (TKIs) in advanced disease. Recently, a newer generation EGFR-TKI, osimertinib, has been approved as an adjuvant treatment for early-stage EGFR-mutant lung cancer after curative resection [6]. Furthermore, novel combinational approaches, such as combining EGFR-TKIs with chemotherapy or an EGFR mesenchymal-epithelial transition factor (MET) bispecific antibody, have recently shown better clinical efficacy than EGFR-TKI monotherapy and emerged as new frontline treatment options [7,8]. However, resistance to TKIs eventually develops via conformational changes in target proteins or activation of bypass signaling pathways [9,10].
The tumor microenvironment (TME) plays a critical role in carcinogenesis and treatment of lung cancer, influencing various aspects of tumor development, progression, and therapeutic outcomes. In lung cancer carcinogenesis, the TME contributes significantly to chronic inflammation, inducing tissue injury and repair cycles [11]. This inflammatory microenvironment promotes bronchioalveolar stem cell growth and activates key signaling pathways such as nuclear factor-kappa B and signal transducer and activator of transcription 3 (STAT3), which are essential for lung cancer development [11,12]. The TME also facilitates immune escape, tumor angiogenesis, and epithelial-mesenchymal transition (EMT), which contribute to cancer progression and metastasis [12,13]. In addition, hypoxia and nutrient deprivation in the TME promote cancer progression by activating angiogenesis, inducing resistance to apoptosis, and selecting resistant clones [14]. Complex interactions between tumors and various cell types, such as macrophages, neutrophils, and cancer-associated fibroblasts (CAFs), significantly affect the clinical outcomes of cancer treatment, particularly immunotherapy [14,15,16,17]. Although the clinical implications of TME in oncogene-addicted lung cancers have not been fully understood, recent evidence demonstrates that those tumors actively interact with the surrounding TME. Further, the TME is influenced by TKI treatment, which may be associated with the development of drug resistance [18]. In this review, we describe the roles of various TME components, such as immune cells, fibroblasts, blood vessels, immune checkpoint proteins, and cytokines, and their involvement in promoting resistance to TKIs in EGFR-mutant lung cancer. We also discuss the potential strategies to overcome TME-associated resistance and the rationale for novel treatments targeting TME or using immunomodulation.
Table 1. Current FDA-approved targeted therapy in advanced NSCLC.
Table 1. Current FDA-approved targeted therapy in advanced NSCLC.
Tyrosine Kinase InhibitorTherapeutic TargetReference
GefitinibEGFR (Exon 19 deletions, L858R mutations)[19]
ErlotinibEGFR (Exon 19 deletions, L858R mutations)[20]
AfatinibEGFR (Exon 19 deletions, L858R mutations, uncommon mutations like G719X, L861Q, S768I)[21]
OsimertinibEGFR (Exon 19 deletions, L858R mutations, T790M)[22]
LazertinibEGFR (Exon 19 deletions, L858R mutations, T790M)[23]
DacomitinibEGFR[24]
AlectinibALK[25]
CeritinibALK, ROS1[26]
BrigatinibALK[27]
LorlatinibALK, ROS1[28]
CrizotinibALK, ROS1, MET[29]
CapmatinibMET exon 14 skipping mutations[30]
TepotinibMET exon 14 skipping mutations[31]
SelpercatinibRET[32]
PralsetinibRET[33]
AmivantamabEGFR, MET[34]
MobocertinibEGFR exon 20 insertion mutations[35]
Dabrafenib + TrametinibBRAF V600E[36]
Encorafenib + BinimetinibBRAF V600E[37]
Abbreviations: EGFR, epidermal growth factor receptor; ALK, anaplastic lymphoma kinase; ROS1, c-ros oncogene 1; MET, mesenchymal-epithelial transition factor; RET, rearranged during transfection; BRAF, v-Raf murine sarcoma viral oncogene homolog B; V600E, valine-to-glutamate substitution at codon 600.

2. Mechanism of Resistance to EGFR-TKIs: An Overview

The emergence of resistance to EGFR TKIs poses a significant challenge in the treatment of EGFR-mutant NSCLC. EGFR mutations play a critical role in the pathogenesis of NSCLC by driving aberrant activation of tyrosine kinase signaling pathways. These mutations lead to the constitutive activation of the receptor’s intrinsic tyrosine kinase domain, independent of ligand binding [38]. The sustained activation leads to the activation of the MAPK, AKT, STAT3, and other downstream oncogenic signaling pathways, promoting processes such as uncontrolled cell proliferation, inhibition of apoptosis, angiogenesis, and metastasis [39]. EGFR-TKI resistance can be categorized into two types, i.e., primary and secondary (acquired) resistance. Primary resistance occurs in patients who do not respond to EGFR-TKIs during frontline treatment or experience early relapse within 6 months. This can be further divided into intrinsic and late primary resistance [40]. The mechanisms underlying primary resistance are not fully understood but may involve pre-existing genomic alterations, including de novo T790M and TP53 mutations, and MET amplification [40].
Secondary or acquired resistance typically develops after an initial response to EGFR-TKIs. This type of resistance develops via either on-target or off-target mechanisms. On-target resistance mechanisms include alterations in the target enzyme, typically the TKI-binding tyrosine kinase, which results in a reduction in the binding affinity of TKIs, making them less capable of inhibiting kinase activity [41]. The most frequent form of on-target resistance is the acquisition of secondary mutations in the gene encoding the target kinase, e.g., the T790M or C797X mutations [42]. For example, T790M mutations alter the configuration of the ATP-binding site of the tyrosine kinase domain, reducing the binding affinity of TKIs and accounting for approximately 50% of the acquired resistance after first- and second-generation EGFR-TKI treatment [43].
Off-target resistance involves changes in cellular pathways or molecules other than the primary target of TKIs, accounting for 10–15% and 30% resistance to early- and third-generation EGFR-TKIs, respectively [44,45,46,47]. These mechanisms include activation of alternative pathways (such as c-MET and AXL), aberrations in downstream pathways (such as phosphatase and tensin homolog loss), and TKI-mediated apoptosis impairment (such as Bcl-2–like 11 (BIM) deletion or polymorphism) [48]. In addition, histological transformations to squamous cell or small cell lung cancer and EMT are also rare but clinically significant resistance mechanisms.

3. Current Treatment Strategy for Overcoming EGFR TKI Resistance with Targetable Co-Mutations

The current approach to overcoming EGFR TKI resistance emphasizes managing actionable co-mutations. Extensive clinical efforts aim to identify these alterations at the time of resistance. However, for patients without detectable genetic changes that allow for targeted intervention, treatment options remain limited. Some key genetic alterations that serve as targets for overcoming EGFR TKI resistance will be discussed here.
  • C797X mutation
Upon progression on osimertinib, approximately 15% of tumors develop on-target mutations [49], with EGFR C797X in exon 20 being the most prevalent. This mutation hampers the covalent binding of osimertinib to the EGFR kinase domain. Other notable acquired mutations include L718Q/V, G719A, and G724S in exon 18 [46]. Fourth-generation EGFR TKIs like BLU-94573 and BBT-17674 have been developed, showing promising initial data. However, further investment in BLU-945 for EGFR-mutant NSCLC has been discontinued, and new treatments are awaited. Additionally, preclinical studies suggest that cancer cells with the acquired C797S mutation after osimertinib therapy remain sensitive to 1G or 2G EGFR TKIs [50]. A multicenter, open-label, phase 1/2 trial (NCT05394831) is investigating JIN-A02, a fourth-generation EGFR-TKI. The results may provide insights into its potential as a treatment option for advanced NSCLC patients with C797S and/or T790M mutations [51]. Another fourth-generation TKI, BDTX-1535, showed promising outcomes in patients with refractory or relapsed EGFR-mutant NSCLC in a phase 2 trial (NCT05256290) [52].
  • MET alteration
In NSCLC, MET-dependent resistance emerges as a significant obstacle, often activated by the formation of homodimers or through trans-activation by other tyrosine kinase receptors. MET amplification contributes to resistance in approximately 50–60% of cases treated with first- and second-generation EGFR TKIs [53,54], and in 15–19% of cases involving third-generation EGFR TKIs [55]. Overcoming this resistance requires the concurrent targeting of both EGFR and MET receptors, highlighting the potential utility of anti-MET agents in combination with EGFR TKIs to achieve a more effective antitumor response [56].
To overcome EGFR TKI resistance combined with MET amplification, a combinatorial approach of EGFR TKI and crizotinib has been attempted [57,58]. Given the limited antitumor efficacy of MET TKIs as monotherapy (ORR: 8.3%) in addressing acquired MET amplification, the combination of a MET TKI with an EGFR TKI has emerged as the most effective strategy to date [59]. This dual inhibition strategy, combined with osimertininb, is being explored in trials such as SAVANNAH (savolitinib) [60], INSIGHT2 (tepotinib) [61], and ORCHARD (savolitinib) [62], which have reported ORRs up to 50% and a median PFS of 5.0 months. Although this approach might improve outcomes compared to standard platinum-pemetrexed chemotherapy [63], its efficacy needs confirmation through ongoing trials like GEOMETRY-E (NCT04816214) and SAFFRON (NCT05261399). For osimertinib-relapsed, chemotherapy-naïve EGFR-mutant NSCLC patients, the combination of amivantamab and lazertinib has shown promising results, especially in tumors with MET overexpression by immunohistochemistry [64]. A recent update from the INSIGHT-2 study (NCT03940703), an open-label, phase 2 trial, reported that the combination therapy of tepotinib 500 mg and osimertinib 80 mg daily achieved a significant ORR of 50.0% (95% CI 39.7–60.3) in patients with MET amplifications who had progressed following initial osimertinib treatment [65].
  • HER2 and HER3 alterations
Human epidermal growth factor receptor 2 (HER2) is a tyrosine kinase receptor encoded by the ERBB2 gene [66]. HER2 amplification is observed in 5% of patients who develop resistance to second-line osimertinib and in 2% of cases using first-line osimertinib [67,68]. Distinct from HER2 amplification, HER2 mutations are considered to be more relevant to lung carcinogenesis, and are detected in approximately 2–4% of patients with NSCLC patients [69,70]. Furthermore, both HER2 amplification and mutation are usually mutually exclusive with other targetable mutations [71,72].
One approach to overcoming HER2 amplification implicated in osimertinib resistance in EGFR-mutant NSCLC involves the combination of trastuzumab-emtansine and osimertinib. In a multicenter, single-arm, phase 1–2 study (NCT03784599), patients treated with this regimen showed a limited ORR of 4% and a median progression-free survival (PFS) of 2.8 months, indicating the need for alternative strategies [73].
HER3 is a member of the EGFR family, and heterodimerizes with other HER proteins. It is involved in cancer cell proliferation by downstream signaling through the PI3K/protein kinase B (AKT) pathway [74]. HER3, when combined with receptors such as MET, HER2, and EGFR, becomes a potent signaling entity, producing strong growth signals that can enhance resistance to targeted therapeutic interventions [75].
An antibody-drug conjugate (ADC) is a class of drug that consists of a monoclonal antibody linked to a cytotoxic payload via a stable chemical linker, enabling targeted delivery of the drug to cancer cells [76]. Initial studies indicate significant potential for ADCs in treating osimertinib-resistant EGFR-mutant NSCLC. This includes ADCs targeting HER3, such as patritumab deruxtecan and BL-B01D1 (a bispecific ADC targeting EGFR and HER3), ADCs targeting TROP2 like datopotamab deruxtecan, and ADCs targeting cMET such as telisotuzumab vedotin [77,78,79,80]. The HERTHENA-Lung02 study (NCT05338970), which included about 560 patients with EGFR mutations who had progressed during EGFR TKI therapy, indicated that patritumab deruxtecan might be effective in treating the EGFR TKI-resistant subgroup [81]. In contrast to the other three agents, telisotuzumab vedotin requires high MET expression for efficacy, defined as a c-Met expression level of 3+ in at least 25% of tumor cells [80].
Other targetable mutations (RET, BRAF, PIK3CA)
RET fusions are also reported as acquired resistance mechanism after EGFR TKI treatment [82,83]. When compared to primary RET fusions, the proportion of CCDC6-RET in patients with acquired resistance to EGFR TKIs was higher. Additionally, RET fusions were more frequently linked to acquired resistance to third-generation EGFR-TKIs compared to earlier generations [84]. Fourteen patients who showed acquired RET fusions after osimertinib treatment underwent osimertinib and selpercatinib, showing modest response rate, disease control rate, and median treatment duration, were recorded at 50% (95% CI: 25–75%, n = 12), 83% (95% CI: 55–95%), and 7.9 months, respectively [85]. For BRAF V600E-mediated osimertinib resistance, combinations such as dabrafenib and trametinib with osimertinib, as well as vemurafenib with osimertinib, have been reported in case studies involving patients resistant to osimertinib [86,87].
Recent studies indicate that the activation of the PI3K/AKT/mTOR signaling pathway contributes to the aggressive nature of lung cancer [88]. Currently, there are no established treatments that effectively target both the EGFR and PI3K/AKT/mTOR pathways simultaneously. Combinatorial approaches such as PI3K inhibitors with EGFR TKIs and PI3K/mTOR inhibitors with EGFR TKIs in overcoming EGFR TKI resistance mediated by the PI3K/AKT/mTOR pathway are to be investigated [89].

4. Role of the TME in EGFR-TKI Resistance

The TME is a dynamic and intricate system consisting of tumor cells, adjacent immune and non-immune cells, and non-cellular components [90]. According to the traditional ‘seed and soil’ theory, tumor cells require a suitable surrounding environment. To escape immune surveillance and survive, tumor cells utilize immune cells and non-cellular components to create an immunosuppressive environment [91]. In addition, the TME is further remodeled during TKI treatment through various components such as tumor-infiltrating immune cells, immune-modulating molecules, cytokines, and chemokines, which are collectively associated with the development of drug resistance [92]. Owing to the critical role of the TME in determining immunotherapy efficacy, it is necessary to understand the important modifications within the TME after TKI treatment, and how these changes contribute to TKI resistance [92] (Table 2, Figure 1). Furthermore, modifying components of the TME can help restore drug sensitivity, delay resistance, and enhance treatment efficacy [93]. The TME, composed of immune cells, stromal cells, and extracellular matrix (ECM) components, plays a critical role in tumor progression and therapeutic response. For instance, targeting immune-suppressive factors such as tumor-associated macrophages (TAMs) or regulatory T cells can improve the effectiveness of targeted therapies and even synergize with immunotherapy [94]. Additionally, tumor modulation strategies allow for the co-targeting of alternative pathways that contribute to resistance against EGFR inhibitors.
Table 2. Explanation of each component of the TME in EGFR-mutant NSCLC.
Table 2. Explanation of each component of the TME in EGFR-mutant NSCLC.
ComponentsEGFR-Mutant StatePotential Role as a Biomarker and Strategic ApproachesReferences
CD8+ lymphocytesCD8+ T-cell infiltration increases in EGFR-mutant NSCLC responsive to TKI but decreases in resistant tumors.Lower levels of CD8+T-cell activity are linked to resistance in EGFR-mutant NSCLC.[95,96,97,98,99,100,101]
CD4+ lymphocytesRegulatory T (FOXP3+) cells (Tregs) are increased in EGFR-mutant NSCLC, contributing to an immunosuppressive microenvironment.High Treg (FOXP3+) counts in EGFR TKI–resistant tumors are linked to immune suppression and resistance.[98,102,103,104,105,106,107]
PD-(L)1PD-L1 expression often increases in EGFR TKI–resistant NSCLC, contributing to immune escape.Elevated PD-L1 levels after EGFR TKI resistance can indicate worse prognosis.
Targeting PD-L1 or related pathways (STAT3, ERK1/2) may enhance response in EGFR TKI–resistant NSCLC.
[98,108,109,110,111,112,113,114,115,116,117,118]
TAMIn EGFR-mutant NSCLC, M2-like macrophages support tumor progression, increasing in TKI-resistant tumors.Increased M2 macrophages are linked to EGFR-TKI resistance. Macrophage polarization (M2 to M1) may predict improved responses in EGFR-TKI-resistant NSCLC.
Combining STING agonists or liposome therapies with EGFR TKIs reprograms TAMs, enhancing tumor regression.
[100,103,119,120,121,122,123]
CytokinesIncreased levels of cytokines such as IL-6, IL-8, and TGF-β promote EGFR TKI resistance and tumor progression.Elevated cytokines (IL-22, IL-6) in plasma and tumor tissue are associated with resistance to EGFR-TKI therapy.
Targeting cytokines (IL-6, IL-8, TGF-β) combined with EGFR-TKIs may help overcome resistance in NSCLC.
[92,93,124,125,126,127,128,129,130,131]
ExosomesExosomes from EGFR-mutant NSCLC carry tumor-related RNA, contributing to EGFR-TKI resistance through signaling pathways.Exosomal miRNA and EGFR expression serve as potential biomarkers for predicting resistance.
Modulating exosomal signaling pathways such as PI3K/AKT and ERK1/2 may overcome TKI resistance.
[103,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147]
CAFCAFs drive resistance in EGFR-mutant NSCLC by promoting EMT and secreting resistance-inducing factors such as HGF, IL-6, and kynurenine, activating pro-survival pathways in cancer cells.CAF markers such as α-SMA, HGF, and podoplanin in tumor tissues could predict EGFR-TKI resistance.
Targeting CAF-derived factors such as HGF or using antifibrotic agents may counteract TKI resistance.
[148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163]
Vasculature (VEGF)VEGF upregulation in EGFR-mutant NSCLC cells contributes to TKI resistance, promoting angiogenesis and tumor progression.High VEGF/VEGFR-2 expression correlates with poor outcomes in EGFR-mutant NSCLC.
Combining VEGF inhibitors such as bevacizumab or ramucirumab with EGFR-TKIs significantly improves PFS.
Anlotinib has shown improved survival outcomes in TKI-resistant patients when combined with immune checkpoint inhibitors.
[164,165,166,167,168]
Abbreviations: EGFR: epidermal growth factor receptor; NSCLC: non-small cell lung cancer; TKI: tyrosine kinase inhibitor; TME: tumor microenvironment; CD8+: cluster of differentiation 8; CD4+: cluster of differentiation 4; PD-1: programmed death-1; PD-L1: programmed death-ligand 1; TAM: tumor-associated macrophage; M1: classically activated macrophage (anti-tumor); M2: alternatively activated macrophage (pro-tumor); STING: stimulator of interferon genes; IL-6: interleukin 6; IL-8: interleukin 8; TGF-β: transforming growth factor beta; CAF: cancer-associated fibroblast; HGF: hepatocyte growth factor; α-SMA: alpha smooth muscle actin; EMT: epithelial-to-mesenchymal transition; VEGF: vascular endothelial growth factor; VEGFR: vascular endothelial growth factor receptor; FGFR: fibroblast growth factor receptor; PDGFR: platelet-derived growth factor receptor; PFS: progression-free survival; EOMES: eomesodermin.

4.1. Cellular Components

4.1.1. Cluster of Differentiation 8 (CD8+) Lymphocytes

CD8+ T-cells exert direct cytotoxic effects on cancer cells by secreting granzymes and perforins. They are activated by tumor-resident antigen-presenting cells or MHC class I molecules on tumor cells, leading to tumor cell destruction and neoantigen release, which can initiate secondary immune responses [95,96].
Fang et al. reported that TKI treatments significantly enhanced immune cell infiltration and cytotoxic activity in samples responsive to TKI, while no such improvements were observed in samples resistant to the therapy [97]. Another study using paired baseline and re-biopsy samples after progression from EGFR-TKI therapy showed that CD8+ and FoxP3+ tumor-infiltrating lymphocyte densities decreased following EGFR-TKI treatment [98]. Another study reported a significant decrease in the number of CD8+ T-cells in tumors after the development of resistance to EGFR-TKIs [99]. In addition, single-cell RNA sequencing revealed that increased infiltration of CD8+ T-cells correlated with a positive response to EGFR-TKI treatment, whereas decreased infiltration correlated with treatment resistance [100].
Although an overall reduction in CD8+ T-cell infiltration has been linked to EGFR-TKI resistance, a specific subset of CD8+ T-cells may be correlated with this resistance. Notably, an increase in eomesodermin (EOMES)-positive CD8+ T-cells, a marker associated with the thymic precursors of self-specific memory-phenotype CD8+ T-cells and immune homeostasis, has been linked to EGFR-TKI resistance [169]. Further analysis confirmed elevated EOMES+ CD8+ T-cell counts in both the tissue and peripheral blood from patients exhibiting resistance to TKIs [169].

4.1.2. CD4+ Lymphocytes

CD4+ T-cells play a central role in coordinating adaptive immune responses by facilitating the recruitment and activation of dendritic cells, crucial for CD8+ T-cell priming [102]. Five main subsets of CD4+ T helper cells have been identified: Th1, Th2, Th17, regulatory T-cells (Treg), and follicular helper T-cells [103,104].
Among various subsets of CD4+ cells, the role of Tregs in EGFR-TKI resistance is relatively well-known. Tregs, generally characterized by the expression of the transcription factor FoxP3, play an important role in maintaining immune tolerance and modulating the immune response [105]. The TME of EGFR-mutant tumors is more likely to have an increased count of Treg cells than that of the wild type [106]. An in vitro study using the EGFR-mutant lung cancer cell lines HCC827 and H4006 co-cultured with activated peripheral blood mononuclear cells and treated with EGFR-TKIs showed notable changes in immune cell dynamics. Treg proportions increased significantly after co-culture and remained unchanged during EGFR-TKI treatment, suggesting their potential role in TKI resistance [170]. In a study using paired tumor samples, the density of Tregs shifted from negative or low at baseline to high after the development of EGFR-TKI resistance in programmed death-ligand 1 (PD-L1)-negative or -positive tumors, indicating an immunosuppressive role of Tregs in oncogene-driven tumors [98].

4.1.3. Tumor-Associated Macrophages (TAMs)

Macrophages are the most abundant innate immune cells in the TME and are commonly referred to as TAMs. The phenotypes and functions of TAMs are heterogeneous and can change dynamically within the surrounding microenvironment. Unpolarized M0 macrophages can be polarized into two functional states: pro-inflammatory M1 macrophages (classically activated) and pro-tumorigenic M2 macrophages (alternatively activated) [171]. These subtypes exhibit high plasticity, allowing them to switch between different forms or alter their functions in the TME [171,172]. M1 macrophages typically exert anti-tumor functions, including direct cytotoxicity and antibody-dependent cell-mediated cytotoxicity to kill tumor cells [173]. In contrast, M2 macrophages promote tumor occurrence, metastasis, angiogenesis, and progression while inhibiting T-cell-mediated anti-tumor immune responses by secreting anti-inflammatory cytokines and immune-suppressive factors [173,174]. In addition, a retrospective study indicated that increased TAM infiltration into tumors was associated with worse clinical outcomes in EGFR-mutant patients treated with EGFR-TKIs [175],
A preclinical study using functional mouse models demonstrated that TAM influx into the lungs precedes the development of a progenitor-like cell state in EGFR-mutant lung alveolar type II epithelial cells, suggesting a potential link between macrophage activity and tumorigenesis in EGFR-mutant cells [120]. A clinical study, using single-cell RNA sequencing of biopsy specimens from EGFR-mutant NSCLC, showed that macrophage infiltration decreased with EGFR-TKI treatment, to which the tumor was sensitive and increased in resistant cases. Upon disease progression, the infiltration of IDO1+ macrophages, proliferative regulatory T-cells, and dysfunctional T-cells, which are scarcely observed in residual diseases, was noted. In contrast, the absence of a TKI response was marked by an increase in immunosuppressive M2-like macrophages [100]. This was further supported by a study by Han et al., in which tissues resistant to osimertinib exhibited an increase in M2 macrophages, indicating enhanced macrophage polarization [103]. In this study, M2 polarization was mediated by tumor-derived exosomes, highlighting the critical role played by exosomes in the development of TKI resistance.

4.1.4. CAFs

CAFs are the primary producers of collagen and directly interact with various stromal cells, including endothelial and inflammatory cells. CAFs have been attributed to several cell types, including locally infiltrating fibroblasts, endothelial cells, pericytes, vascular adventitial fibroblasts, and cancer cells that undergo fibroblastic changes during EMT [148]. Multiple markers, such as α-smooth muscle actin (α-SMA), tenascin-C, platelet-derived growth factor receptor (PDGFR)-A/B, fibroblast activation protein-α, CD90, and podoplanin, are used to identify CAFs. Although α-SMA-positive fibroblasts are commonly known as myofibroblasts, all CAFs do not express α-SMA [148].
As essential stromal components of the TME [149], CAFs play a central role in the development of EGFR-TKI resistance. When EGFR TKI-sensitive NSCLC cells are co-cultured with CAFs, the cells become resistant to TKIs, suggesting that resistance may develop through direct contact [150]. CAFs significantly contribute to lung cancer resistance to EGFR-TKIs by inducing EMT through specific CAF-mediated signaling pathways [151]. CAFs are heterogeneous, with various subtypes distinguished by markers such as vimentin, α-SMA, podoplanin, and periostin [152,153].
In addition, CAFs produce humoral factors that contribute to EGFR-TKI resistance. Suzuki et al. identified CAF-derived hepatocyte growth factor (HGF) as an important factor in inducing EGFR-TKI resistance. A study using an EGFR-mutant cell line PC-9, treated with conditioned media from CAFs and their matched non-cancerous tissue-associated fibroblasts, suggested that EGFR-TKI resistance-promoting CAFs are marked by the elevated secretion levels of HGF. This was demonstrated by the effects observed with the use of an HGF-neutralizing antibody [154]. In a previous study involving mouse models and human samples, proteins produced by CAFs from tumor tissues, such as kynurenine, activated the downstream AKT and extracellular signal-regulated kinase (ERK) pathways, contributing to EGFR-TKI resistance [155]. In a study by Tan et al., a lung-on-a-chip model was employed to co-culture NSCLC cell lines with human fibroblasts and endothelial cells to create a simulated TME for examining EGFR-TKI resistance mechanisms. Human fetal lung fibroblasts transform into CAFs and IL-6 facilitates this transformation, inducing EMT in NSCLC cells, which contributes to their resistance to osimertinib [156].
Evidence suggested that certain CAF subtypes play a dominant role in resistance to EGFR-TKIs. In a study using a xenograft model, a CAF subpopulation enriched in IL-6 production was the predominant CAF type in drug-tolerant persister cells that emerged after repeated erlotinib treatment [157]. Additionally, podoplanin-positive CAFs have been implicated in EGFR TKI resistance in EGFR-mutant lung adenocarcinoma, as evidenced by in vitro studies showing that cancer cells co-cultured with these CAFs exhibit increased resistance [150]. Moreover, adenocarcinomas with solid-predominant histology, which tend to exhibit high primary resistance to EGFR-TKIs, frequently contain more podoplanin-positive CAFs than other histological variants [158,159]. In contrast, a study using patient-derived CAFs demonstrated that CAF heterogeneity contributes to overall TKI resistance. We found that MET and/or fibroblast growth factor receptor (FGFR) activation by CAFs could repeatedly rescue EGFR-mutant cancers [160].

4.2. Non-Cellular Components

4.2.1. Cytokines

Cytokines are crucial TME components and activate or suppress immune responses against cancer cells. The cytokine levels in the TME are also affected by EGFR-TKI treatment [92]. Generally, immunosuppressive cytokines are upregulated, whereas immune-promoting factors are downregulated in EGFR-mutant tumors [92]. Evidence has consistently reported that elevated cytokine levels in the plasma and/or tumor samples are associated with TKI resistance.
In one study, IL-22 levels were higher in the EGFR TKI-resistant group than in the EGFR TKI-sensitive group. These levels were also associated with EGFR TKI efficacy [124]. Another study revealed that elevated CXCL10 levels during early EGFR-TKI treatment enhance oncogenic signaling and promote EGFR-TKI resistance through autocrine and paracrine pathways [125].
IL-6 is a known activator of the Janus kinase/STAT3 pathway and is involved in lung cancer metastasis [176]. In a study involving osimertinib-resistant cell lines and xenograft models, IL-6 levels were elevated. This study further demonstrated that IL-6 inhibition could reverse this resistance [126]. In a study using a genetically engineered mouse model of EGFR-mutant NSCLC, tumors resistant to EGFR-TKIs showed a notable increase in IL-6 secretion. The reduction in IL-6 was associated with an increased activity of infiltrating natural killer (NK) and T-cells and a decrease in Treg and Th17 cell populations [127].
IL-8 contributes to angiogenesis, cancer cell growth and survival, tumor cell motility, and leukocyte infiltration, which promote tumor progression [177]. In NSCLC, elevated IL-8 levels are associated with treatment resistance, with data from The Cancer Genome Atlas (TCGA) dataset showing a negative prognostic impact of high IL-8 levels in lung cancer, particularly in patients treated with chemotherapy and/or immunotherapy [128,178]. In EGFR-mutant lung cancer, IL-8 is upregulated in gefitinib-resistant cells, and high plasma IL-8 levels are correlated with shorter progression-free survival (PFS) in patients treated with EGFR-TKIs [179]. Moreover, IL-8 suppression enhanced gefitinib-induced cell death in gefitinib-resistant cells by inducing the loss of stem cell-like characteristics, suggesting a potential role of IL-8 in EGFR-TKI resistance, consistent with chemotherapy and immunotherapy findings [179]. In another study, blocking IL-8 signaling effectively reduced the mesenchymal features of TKI-resistant cells and markedly enhanced their susceptibility to erlotinib, verifying that IL-8 is a potential therapeutic target for overcoming EGFR-TKI resistance [129].
Transforming growth factor-beta (TGF-β) plays a complex and multifaceted role in cancer development and treatment [130]. TGF-β acts as a tumor suppressor in the early stages of cancer, inhibiting cell growth and promoting apoptosis [180]. However, as tumors progress, it promotes tumor growth, invasion, and metastasis [181]. This dual role of TGF-β complicates its relationship with EGFR-TKI treatment, as the cancer stage and specific tissue context can influence its effects. The TGF-β signaling pathway may contribute to TKI resistance by promoting tumor cell survival and metastasis in the later stages of cancer [181]. Additionally, both EGFR and TGF-β pathways are involved in regulating the TME and immune responses, which can impact treatment efficacy [180,182]. TGF-β2 was upregulated in osimertinib-resistant cells, leading to EMT and mothers against decapentaplegic homolog 2 pathway activation [131].

4.2.2. Exosomes

Exosomes are extracellular vesicles, 30–150 nm in diameter, produced via the endoplasmic reticulum pathway. They function as carriers, transporting an array of substances such as proteins, nucleic acids, metabolites, and lipids originating from diverse cells [132], and play a pivotal role in cell-to-cell communication [133,134]. Lung cancer cells produce significant amounts of extracellular vesicles, predominantly as exosomes [135]. Normal human blood contains approximately 2000 trillion exosomes; however, this number is significantly increased in patients with cancer, with counts reaching approximately 4000 trillion [136]. These exosomes play crucial roles in various immune functions, including antigen presentation, immune activation and suppression, surveillance, and cell communication [137].
Especially, exosomal miRNAs may also play an important role in predicting treatment resistance and modulating the TME. The miRNA content in circulating exosomes closely resembles that in the originating cancer cells, suggesting their potential application in cancer diagnostics [140]. Furthermore, detecting exosomal miRNAs in body fluids underscores their promising role as disease biomarkers to predict treatment resistance [141]. miRNAs, abundantly present in exosomes, can modulate the stability or translation of target mRNAs, significantly altering the TME in lung cancer [142]. Liu et al. revealed that treatment with exosomes released by EGFR TKI-resistant lung cancer cells resulted in the acquisition of resistance in TKI-sensitive cells via phosphoinositide 3-kinase (PI3K)/AKT signaling pathway activation, suggesting a potential contribution of exosomes to the emergence of TKI resistance [143].
Notably, exosomes are critically involved in the polarization of macrophages into the M2 phenotype, which plays an immunosuppressive role in the TME [103]. Additionally, exosomes extracted from TAMs promote TKI resistance in lung cancer cells through AKT, ERK1/2, and STAT3 signaling pathway reactivation [144]. In a study evaluating the mechanism of action of exosome-derived miRNAs in osimertinib resistance, exosome-derived miR-184 and miR-3913-5p expression was significantly increased in the blood of patients with osimertinib resistance, suggesting that specific miRNA signatures are involved in the development of TKI resistance and can be used to predict resistance [145].

4.2.3. VEGF and Vasculature

Angiogenesis is a key feature of tumors as it helps provide a continuous supply of nutrients. This process is controlled by several growth factors, with VEGF as a key mediator [164,165]. VEGF is often upregulated in response to hypoxic conditions within tumors [183,184], stimulating endothelial cell proliferation and new capillary formation to meet the metabolic demands of expanding cancer cells [185,186]. Interestingly, VEGF shares common downstream signaling with the EGFR pathway and activated EGFR signaling can upregulate VEGF expression [187]. Furthermore, a preclinical study showed that VEGF levels were significantly higher in EGFR-mutant cells than in wild-type cells [188].
In addition to its role in cancer cell proliferation, VEGF-driven angiogenesis contributes to an immunosuppressive TME [189,190]. Increased VEGF expression facilitates the recruitment of immune cells, such as TAMs and Tregs, which negatively regulate anti-tumor immune responses [191,192]. VEGF-mediated modulation also increases vascular permeability, creating a leaky and abnormal vasculature that limits the effective penetration of immune cells and therapeutic agents into tumors [193]. Current therapies targeting the VEGF/VEGFR pathway, including VEGF or VEGFR neutralizing antibodies and TKIs, have demonstrated substantial efficacy in patients with NSCLC [194].

4.2.4. PD-L1 Expression

PD-1 is a key immune checkpoint that facilitates cancer cell immune evasion by interacting with PD-L1 on tumor cells, ultimately suppressing the tumor-killing effects of CD8+ T-cells [108]. High PD-L1 expression is a well-known predictor for excellent clinical outcomes with anti-PD-(L)1 treatment, although it is not an optimal predictive biomarker. Notably, EGFR-mutant lung cancer is characterized by an uninflamed phenotype with a high frequency of inactive tumor-infiltrating lymphocytes (TIL)s, low PD-L1 expression, and low tumor mutation burden [195,196,197,198]. In contrast to immunotherapy, studies have consistently revealed that high PD-L1 expression in the tumor tissues is associated with TKI resistance and poor clinical outcomes [198,199,200]. Interestingly, TKI treatment may affect PD-L1 expression, and the upregulation of PD-L1 has been consistently reported during the emergence of TKI resistance.
In a study on 138 patients with EGFR mutation who underwent re-biopsy after progression following EGFR-TKI failure, the proportion of patients with ≥50% PD-L1 expression increased significantly from baseline (14%) to 28% [98]. Similar results have been reported in other studies [111,112]. Various mechanisms have been suggested for PD-L1 expression elevation in EGFR TKI resistance. A study using TCGA database and paired NSCLC samples suggested that HGF, c-MET amplification, and the T790M mutation are involved in the upregulation of PD-L1 in NSCLC [109]. An in vitro study reported that continuous TKI treatment resulted in PD-L1 overexpression, which correlated with T-cell proliferation suppression and STAT3 and ERK1/2 pathway activation [115]. Another study using next-generation sequencing showed that mutations in the PI3K signaling pathway may lead to initial resistance to EGFR-TKIs with elevated PD-L1 levels [116]. Additionally, AKT–mammalian targeting of rapamycin pathway activation and increased BIM expression have been suggested as possible mechanisms for increased PD-L1 expression in EGFR-mutant NSCLC [117,118].

4.2.5. Extracellular Matrix (ECM)

Studies emphasize the significant influence of the ECM in driving resistance to EGFR TKIs in EGFR-mutant NSCLC [201]. Integrins, crucial transmembrane receptors interacting with ECM components, play an essential role in activating signaling pathways linked to therapeutic resistance [202,203]. Integrin β1, for instance, engages with collagen-rich ECM to promote cell adhesion and trigger alternative signaling cascades such as FAK/Src and PI3K/AKT, allowing tumor cells to proliferate [204]. Similarly, integrin β3 has been associated with acquired resistance to EGFR TKIs, with its upregulation observed in resistant tumors and its inhibition being explored as a strategy to restore TKI sensitivity [205]. Collagen, a fundamental ECM component, further contributes to resistance by modifying tumor stiffness and interstitial pressure, ultimately hindering drug penetration [201]. Moreover, collagen type I has been shown to induce resistance by activating mTOR signaling through an Akt-independent mechanism, fostering the survival and proliferation of EGFR-mutant cancer cells [206].

4.2.6. Adenosine Pathway

Recent studies have highlighted the critical role of the adenosine pathway in immune suppression, particularly in regulating lymphocyte activity. Within the TME, adenosine is a key modulator of immune responses [207]. Its accumulation is facilitated by ectonucleotidase CD73, which promotes adenosine production and subsequently activates immunosuppressive pathways in tumor-infiltrating immune cells [208]. Signaling through the adenosine A2A receptor has been shown to impair the cytotoxic function of CD8+ T cells and natural killer cells while promoting the differentiation of CD4+ T cells into regulatory T cells [209]. Additionally, innate immune cells within the TME express adenosine receptors, and activation of these receptors enhances the immunosuppressive functions of M2 macrophages and amplifies the effects of myeloid-derived suppressor cells [210].
Le et al. report that EGFR-mutated NSCLC, unlike EGFR wild-type, exhibits upregulation of the CD73/adenosine pathway. In the study, coculture systems with EGFR-mutant NSCLC cells demonstrated that CD73 knockdown reduced the proportion of regulatory T cells. Furthermore, treatment with an anti-CD73 antibody resulted in tumor reduction in an EGFR-mutant mouse model [211]. A study by Tu et al. reported that CD73 expression was elevated in EGFR-mutant NSCLC compared to EGFR wild-type tumors and was regulated by EGFR signaling. In a xenograft model, combination treatment with anti-PD-L1 and anti-CD73 antibodies significantly suppressed tumor growth, increased the number of tumor-infiltrating CD8+ T cells, and enhanced IFN-γ and TNF-α production by these T cells [212].

5. Potential Strategies for Overcoming TKI Resistance Through TME Modulation

While the previous sections discussed various TME components in relation to EGFR-TKI resistance, translating this knowledge into clinical practice remains the ultimate objective. In the current treatment setting, the goal of addressing EGFR-TKI-resistant tumors is to immediately identify actionable co-mutations. However, ongoing research on the potentially treatable components of the TME should expand future treatment options. Current research focuses on identifying biomarker candidates, elucidating the roles of targetable co-mutations in TME components, and identifying potential therapeutic targets that can be used to overcome EGFR-TKI resistance.

5.1. Targeting CAFs

CAFs are considered potential targets for treatment because of their role in the pathogenic crosstalk between lung fibrosis and cancer, which contributes to the fibrotic stromal components of the TME [161]. Blocking CAF-mediated signals, depleting CAF populations, and extracellular matrix remodeling may be potential therapeutic strategies for targeting the tumor-promoting functions of CAFs [153].
Preclinical models have explored the ability of antifibrotic agents pirfenidone and nintedanib to inhibit CAFs, which have shown potential as components of combination therapies aimed at slowing cancer progression [162,163]. Interestingly, pirfenidone has immunomodulatory effects as well as an anti-fibrotic effect. Studies have demonstrated that pirfenidone monotherapy can attenuate tumor growth and enhance T-cell-mediated inflammation in tumors. When combined with PD-L1 blockade, it significantly delayed tumor growth and increased survival than either treatment alone. This combination therapy promotes the expression of genes associated with innate and adaptive immune responses, resulting in increased immune cell infiltration and optimal T-cell positioning [213].
Nintedanib is a multi-target TKI that inhibits angiogenesis-related receptors, including VEGF, fibroblast growth factor (FGF), and platelet-derived growth (PDGF) [214,215]. A study demonstrated that nintedanib effectively blocked the ability of both normal and tumor-derived supernatants to facilitate aggressive cancer cell characteristics in lung cancer cell lines [216]. Although the study did not find significant changes in FGF, PDGF, or VEGF levels, it did reveal high levels of HGF, suggesting that HGF potentially facilitates cell migration and proliferation.
However, further studies should determine the potential value of CAF inhibition. One challenge in targeting CAFs is prioritizing specific subtypes within the heterogeneous CAF population and disrupting critical interactions between CAFs and other cells, which, when inhibited, can lead to the regression of TKI-resistant tumor cells.

5.2. Targeting TAMs

The TAM-mediated T-cell suppression is involved in EGFR-TKI resistance [121]. An in vivo study demonstrated that combining a stimulator of interferon genes (STING) agonist with osimertinib to reprogram immunosuppressive TAMs effectively induced regression of advanced tumors [122]. TAM reprogramming has emerged as a promising strategy for cancer treatment, and numerous clinical trials have explored this approach. According to a recent review, approximately 200 TAM-reprogramming agents have been investigated in more than 700 clinical trials [217]. However, strong anti-tumor efficacy is uncommon, suggesting the need to identify biomarkers for eligible patient populations and compare similar treatments earlier in the development process [217].
Reversal of the M2 phenotype to M1 is effective at overcoming gefitinib resistance. Methionine sulfoxide reductase A (MsrA) helps protect the T790M-mutant EGFR protein. When macrophages switch from the M2 to M1 phenotype, the production of reactive oxygen species (ROS) increases, lowering MsrA levels and accelerating EGFR breakdown [123]. Peng et al. developed a trastuzumab-modified liposome carrying gefitinib and vorinostat. Trastuzumab targets HER2-positive NSCLC cells, whereas vorinostat reverses the polarization of tumor-promoting M2 macrophages [123]. Similarly, Yin et al. created PD-L1-modified liposomes that delivered gefitinib and simvastatin, specifically targeting TAMs [218]. These two studies utilizing liposomes to target TAMs showed that converting macrophages from M2 to M1 increased ROS levels, inhibited angiogenesis, and boosted the release of immune-stimulating factors such as TGF-β. Trastuzumab-modified mannosylated liposomes and PD-L1-modified liposomes, both designed to deliver gefitinib, demonstrated strong anti-tumor effects and good safety profiles in NSCLC mouse models with T790M mutation. The results of these two studies indicated that targeting TAMs and modifying the TME could represent potential therapeutic strategies [219].

5.3. Targeting Cytokines

A study on EGFR-mutant cells suggested that combinatorial treatment with cytokines and osimertinib may be a potential treatment strategy. Ding et al. demonstrated that the combination of IL-12 with osimertinib synergistically affected tumor suppression. The authors suggested that this synergistic effect was due to increased immune cell infiltration, elevated secretion of immune-related factors, and reduced levels of immunosuppressive myeloid-derived suppressor cells [93]. However, few studies have provided preclinical or clinical evidence to support the intervention of other cytokines such as IL-6 and IL-8. To date, only a few effective combinatorial treatment strategies that directly target cytokines have been reported. A possible explanation is that compared to wild-type EGFR NSCLC, modulation of the immune microenvironment may not yield favorable outcomes. This is supported by recent studies showing that immunotherapy demonstrated limited efficacy in patients with EGFR-mutant NSCLC who progressed on EGFR-TKI therapy [220].
Although not specifically focused on patients with EGFR mutations, the NCT04691817 trial is currently enrolling participants to evaluate the safety and efficacy of tocilizumab, a monoclonal antibody targeting the IL-6 receptor, in combination with atezolizumab, for the treatment of NSCLC. This phase Ib–II trial is investigating the potential of this combination in patients with locally advanced or metastatic NSCLC who were refractory to first-line immune checkpoint inhibitor (ICI)-based therapy [221].

5.4. Immunotherapy

Increased activity of immune cells showing anti-tumor effects is a potential option to overcome EGFR-TKI resistance. Several key trials have been performed to identify effective immunotherapeutic options for patients with EGFR-mutant NSCLC who experience disease progression after TKI treatment.
The KEYNOTE-789 trial is a randomized, double-blind, phase III study evaluating the efficacy of pembrolizumab in combination with pemetrexed and platinum-based chemotherapy in patients with TKI-resistant, EGFR-mutant, and metastatic non-squamous NSCLC (NCT03515837). Patients with stage IV non-squamous NSCLC, confirmed DEL19 or L858R EGFR mutations, and progression after EGFR-TKI therapy were randomized 1:1 to receive pembrolizumab (200 mg every three weeks for up to 35 cycles) or placebo along with four cycles of pemetrexed and carboplatin or cisplatin, followed by pemetrexed maintenance therapy. In total, 492 patients were enrolled, and at the second interim analysis, the median PFS for pembrolizumab and chemotherapy and placebo and chemotherapy arms were 5.6 and 5.5 months, respectively (hazard ratio [HR] 0.80; 95% confidence interval [CI], 0.65–0.97; p = 0.0122). At the final analysis, the median overall survival (OS) in the pembrolizumab and placebo arms was 15.9 and 14.7 months, respectively (HR 0.84; 95% CI, 0.69–1.02; p = 0.0362). Treatment-related adverse events (TRAEs) of grade ≥ 3 occurred in 43.7% of patients receiving pembrolizumab plus chemotherapy, compared to 38.6% in patients receiving placebo plus chemotherapy. However, the addition of pembrolizumab to chemotherapy did not significantly improve PFS or OS [220]. The ongoing benefits of immunotherapy-based treatments for EGFR-mutant NSCLC following failure of EGFR-TKIs remain uncertain.
However, some studies have indicated potential benefits of immunotherapy-based treatments in patients with pretreated EGFR-mutant NSCLC. Beyond the commonly used immunotherapy options, toripalimab, a humanized IgG4κ monoclonal antibody targeting the PD-1 receptor, has demonstrated efficacy as a second-line treatment for patients with EGFR-mutant advanced NSCLC in a phase II study. The study reported an overall response rate (ORR) of 50.0%, disease control rate of 87.5%, median PFS of 7.0 months, and OS of 23.5 months [222]. In the phase II ATLANTIC trial of durvalumab in heavily pretreated patients with NSCLC, a subgroup analysis was dedicated to those with EGFR- and anaplastic lymphoma kinase (ALK)-positive disease (n = 111). For patients with PD-L1 expression ≥ 25%, the ORR was 12%, with median PFS and OS of 1.9 (95% CI, 1.8–3.6) and 13.3 (95% CI, 6.3–24.5) months, respectively. Among those with PD-L1 expression < 25%, the median PFS and OS were 1.9 (95% CI, 1.8–1.9) and 9.9 (95% CI, 4.2–13.3) months, respectively. Specifically, EGFR-positive patients (n = 97) had a median OS of 16.1 months (95% CI, 6.2–33.2) [223,224]. Additionally, the ORIENT-31 and ATTLAS trials demonstrated that adding a VEGF inhibitor to immunotherapy and chemotherapy regimens significantly improved survival outcomes [225,226].
ICI-based immunotherapy is not considered the standard treatment for treatment-naïve patients with EGFR-mutant NSCLC. The potential benefit of ICI-based therapies in patients with advanced EGFR-mutant NSCLC who have progressed on EGFR-TKIs remains uncertain. This requires further investigation, given the mixed outcomes of studies such as KEYNOTE-789 and phase II ATLANTIC [223,227]. Additionally, owing to the risk of immune-related adverse events (IRAEs) associated with immunotherapy [228,229], a cautious approach is required to minimize unnecessary risks to patients [230]. In this context, identifying the subtypes of EGFR-mutant NSCLC that are likely to benefit from immunotherapy-based treatments after progression to prior TKI therapy is crucial.
Owing to the relatively small number of patients enrolled in studies assessing the efficacy of immunotherapy in patients with EGFR mutations, no definite subgroups that would benefit from immunotherapy have been established. A retrospective study indicated that patients with prior TKI treatment and PFS < 10 months exhibited excellent response to subsequent anti-PD-1/PD-L1 immunotherapy [231]. The type of EGFR mutation and the presence of the T790M mutation were significantly associated with PFS in immunotherapy-based treatments. An indirect comparative meta-analysis showed that patients with the L858R mutation (HR 0.52; 95% CI, 0.37–0.72) and those lacking the T790M mutation (HR 0.50; 95% CI, 0.35–0.71) demonstrated significantly greater benefits from immunotherapy regimens than their counterparts [225]. High PD-L1 expression may not predict a strong response to immunotherapy, as ≥50% patients with PD-L1 expression also showed limited efficacy in a phase II trial (NCT02879994) of pembrolizumab in TKI-naïve patients [232].

5.5. Combination of Anti-Angiogenic and Immunotherapy

As vasculature is a key component of the TME, VEGF, a key mediator, has been targeted for intervention in various solid tumors. Multiple studies have shown that combining a VEGF inhibitor with an EGFR-TKI is significantly more effective than EGFR-TKI monotherapy in patients with advanced EGFR-mutant NSCLC.
A phase III, randomized open-label trial (NEJ026) compared erlotinib with bevacizumab to erlotinib alone in patients with stage IIIB–IV or recurrent non-squamous NSCLC with EGFR mutations. A total of 228 patients were randomly assigned in a 1:1 ratio to receive erlotinib (150 mg daily) plus bevacizumab (15 mg/kg every 21 days) or erlotinib monotherapy. The median follow-up duration was 12.4 months. At the interim analysis, the median PFS in the combination and monotherapy groups were 16.9 and 13.3 months, respectively (HR 0.605; p = 0.016). Serious adverse events occurred in 9% and 5% of patients in the combination and monotherapy groups, respectively [166].
ORIENT-31 is a randomized, double-blind, multicenter, phase III trial evaluating the combination of sintilimab, a PD-1 inhibitor, with the bevacizumab biosimilar IBI305, and chemotherapy in patients with EGFR-mutant non-squamous NSCLC who have progressed after EGFR-TKI therapy. A total of 444 patients who experienced disease progression after TKI were assigned to either the sintilimab plus IBI305 group or the other arms, including chemotherapy. With a median follow-up of 9.8 months (IQR 4.4–13.3), the group receiving sintilimab, IBI305, and chemotherapy showed significantly improved PFS than the chemotherapy-alone group (median PFS, 6.9 months [95% CI, 6.0–9.3] vs. 4.3 months [4.1–5.4]; HR, 0.46 [0.34–0.64]; (p < 0.0001). This combination demonstrated efficacy with a tolerable safety profile [226].
Similar clinical superiority has also been demonstrated with ramucirumab. A double-blind, phase III trial was conducted in patients with EGFR-mutant stage IV NSCLC without central nervous system metastasis (NCT02411448). A total of 449 patients were randomized 1:1 to receive erlotinib (150 mg/day) with intravenous ramucirumab (10 mg/kg) or a placebo every 2 weeks. The PFS was significantly longer in the ramucirumab plus erlotinib group (19.4 months) than in the placebo group (12.4 months). Serious treatment-emergent adverse events occurred in 29% of participants in the ramucirumab group and 21% in the placebo group. The most common serious adverse events were pneumonia (3%), cellulitis, and pneumothorax (2%). These results show a notable improvement in PFS with combination therapy, despite a relatively higher incidence of adverse events [233].
These clinical benefits may be associated with the role of VEGFR-2 in TKI resistance. A study by Osude et al. demonstrated that in EGFR-TKI-resistant NSCLC cell lines—specifically those harboring EGFR double mutations (L858R and T790M)—the expression of VEGF, VEGFR-2, and its co-receptor neuropilin-1 (NP-1) was significantly more elevated than that in parental or single-mutant cells. Immunofluorescence and flow cytometry confirmed increased expression of VEGFR-2 and NP-1 in TKI-resistant cells. Functional assays further revealed that combining a VEGFR-2 inhibitor with erlotinib reduced the viability of EGFR double-mutant NSCLC cells (9%) significantly more than erlotinib alone (72%) [167].
Unlike ramucirumab and bevacizumab, anlotinib inhibits multiple pathways, including VEGFR, FGFR, and PDGFR [168]. A retrospective analysis evaluated the effectiveness of combining anlotinib and ICIs in patients with NSCLC resistant to EGFR-TKI therapy. Eighty patients with lung adenocarcinoma were reviewed, with 38 receiving anlotinib plus ICIs and 42 receiving platinum chemotherapy. After a median follow-up of 15.7 months, the combination group showed significantly better PFS (4.3 vs. 3.6 months; p = 0.005) and OS (14.2 vs. 9.0 months; p = 0.029) than the chemotherapy group. Among those receiving combination therapy, 73.7% received prior treatment, with a disease control rate of 92.1%. Adverse events led to discontinuation in four patients, but most reactions were manageable and reversible [234].
The IMpower150 trial showed significant improvements in PFS and OS with atezolizumab plus bevacizumab plus carboplatin plus paclitaxel (ABCP) versus standard-of-care bevacizumab plus carboplatin plus paclitaxel in chemotherapy-naïve patients with non-squamous NSCLC [235]. In the final analysis for the subgroup with EGFR mutations, the ABCP regimen improved OS in both the overall (HR 0.60; 95% CI, 0.31–1.14) and TKI-treated (HR 0.74; 95% CI: 0.38–1.46) populations [236]. Building on the dual-targeting approach of the PD-L1 and VEGF pathways, the HARMONi-A trial evaluated ivonescimab, a bispecific antibody against PD-1 and VEGF, combined with chemotherapy, and demonstrated that dual inhibition can significantly improve PFS in EGFR-mutant NSCLC after prior TKI therapy. The trial found that the ivonescimab combination notably increased median PFS (7.1 months; 95% CI, 5.9–8.7) more than the placebo (4.8 months; 95% CI, 4.2–5.6; difference of 2.3 months; HR, 0.46 [95% CI, 0.34–0.62]; p < 0.001). The ORR was 50.6% (95% CI, 42.6–58.6%) for ivonescimab versus 35.4% (95% CI, 28.0–43.3%) for the placebo. Another notable finding was the pronounced PFS benefit in the T790M-positive group (HR 0.22; 95% CI, 0.09–0.54). Regarding the safety profile, ≥grade 3 adverse events were reported in 61.5% and 49.1% of patients treated with ivonescimab and placebo, primarily related to chemotherapy [237]. Considering the limitations of monotherapy in EGFR-mutant NSCLC, the addition of VEGF inhibition to immunotherapy may be a potential treatment option for patients who have progressed on EGFR-TKIs.

5.6. Bispecific Antibody Targeting EGFR and MET

Amivantamab is a bispecific antibody that targets both EGFR and MET in NSCLC treatment [238,239]. The unique ability of the drug to bind to two receptors simultaneously sets it apart from traditional antibodies that target only a single receptor [239]. Its mechanism of action involves three primary components: immune cell-mediated killing, receptor internalization and degradation, and the inhibition of ligand binding to both EGFR and MET receptors [240]. Amivantamab plays a significant role in immune modulation by enhancing Fc function. The drug binds tightly to Fcγ3R, mediating macrophage and NK-cell-mediated killing of cancer cells [241]. In vitro studies showed that adding isolated human immune cells notably enhanced amivantamab-mediated EGFR and MET downregulation, resulting in dose-dependent cancer cell death [242]. In this study, monocytes and/or macrophages were critically involved in Fc interaction-mediated EGFR/MET downregulation via trogocytosis [242]. This Fc-dependent macrophage-mediated anti-tumor mechanism represents a novel approach for antibody-based cancer therapies.
Promising results were obtained in a recent phase III MARIPOSA-2 trial that evaluated the efficacy of amivantamab combined with chemotherapy, with or without lazertinib, versus chemotherapy alone in patients with advanced EGFR-mutant NSCLC who experienced disease progression after osimertinib treatment [243]. In this study, PFS improved in patients receiving amivantamab-based regimens significantly more than in those receiving chemotherapy alone. Response rates were also higher with combination therapies, leading to partial or complete responses in > 50% of participants without new safety signals [243]. These results suggest that the amivantamab-based combination offers a new treatment option for overcoming resistance to EGFR-TKIs, highlighting the potential therapeutic role of immune modulation even in oncogene-addicted lung cancer.
Many combinatorial treatments, including various options such as TKIs, target-specific antibodies, VEGF inhibitors, and conventional chemotherapy, should be developed to optimize strategies for overcoming resistance to TKI treatment. Consequently, understanding the TME components in EGFR-mutant NSCLC is becoming increasingly important.

5.7. High-Dose Administration of TKI

One treatment strategy for overcoming TKI resistance is the administration of high-dose TKIs. In EGFR-mutant NSCLC with leptomeningeal metastases, high-dose erlotinib has demonstrated efficacy with a tolerable safety profile [244]. A systematic review evaluating the impact of higher-than-approved TKI dosing on achieving increased maximum plasma concentrations, which may lead to higher intratumoral drug levels and improved efficacy, found that high-dose intermittent TKI treatment regimens can result in elevated plasma concentrations while maintaining tolerable toxicity [245].
However, given the availability of other treatment options, preemptive exposure to doses higher than the approved TKI regimen cannot be considered a priority.

6. Future Perspectives

Numerous studies have emphasized the significance of each TME component in managing EGFR-mutant NSCLC. Various platforms have been used to assess the levels of these components at different transcriptional stages. For clinical implementation, much of the available research is often preliminary and requires further refinement and validation. Owing to the complexity of the TME, an intuitive approach is essential for integrating diverse information. Machine learning has recently been applied to TME analysis, prognosis prediction, and biomarker identification for predicting resistance [246,247].
Employing advanced technology can effectively organize comprehensive genetic, molecular, clinical, and spatial data of the TME, targeting specific clinical objectives, such as predicting and managing EGFR-TKI resistance. Developing a scoring system that weighs multiple clinically significant parameters related to TKI resistance could further refine this approach. Clinicians and researchers should also explore how to incorporate both genetic and non-genetic information from TME components into selecting appropriate treatment strategies. A surge in new treatment regimens has recently been introduced, necessitating a tailored approach to optimal patient care.

7. Conclusions

Various resistance mechanisms and targetable mutations exist in patients with NSCLC who have been treated with TKIs. Understanding the frequent resistance mechanisms specific to each targeted therapy, including those influenced by specific TME components, is essential in overcoming resistance. Additionally, predicting potential resistance to TKIs before treatment can improve patient outcomes.

Author Contributions

Conceptualization, J.U.L. and S.H.L. (Sang Hoon Lee); methodology, J.J., Y.W.K., C.Y.K., S.H.L. (Sang Hoon Lee), D.W.P., S.I.C., W.J. and C.D.Y.; software, J.U.L. and S.H.L. (Seung Hyeun Lee); validation, J.U.L. and S.H.L. (Seung Hyeun Lee); formal analysis, J.U.L. and S.H.L. (Seung Hyeun Lee); investigation, J.U.L. and S.H.L. (Seung Hyeun Lee); resources, J.J., Y.W.K., C.Y.K., S.H.L. (Sang Hoon Lee), D.W.P., S.I.C., W.J. and C.D.Y.; data curation, all authors; writing—original draft preparation, J.U.L. and S.H.L. (Seung Hyeun Lee); writing—review and editing J.U.L. and S.H.L. (Seung Hyeun Lee); visualization J.U.L. and S.H.L. (Seung Hyeun Lee); supervision, J.U.L. and S.H.L. (Seung Hyeun Lee); project administration, J.U.L. and S.H.L. (Seung Hyeun Lee); funding acquisition, J.J. and S.H.L. (Seung Hyeun Lee). All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Science, ICT, and Future Planning (Grant No. 2023R1A2C1003763), and by the Korea Institute of Oriental Medicine (No. KSN2026240).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The datasets used or analyzed in the current study are available from the corresponding author upon reasonable request.

Acknowledgments

During the preparation of this work, the authors used ChatGPT (Model GPT-4o) for language correction. After using these tools/services, the authors reviewed and edited the content as needed and therefore take full responsibility for the content of the publication.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
NSCLCnon-small cell lung cancer
TKItyrosine kinase inhibitor
METmesenchymal-epithelial transition
TMEtumor microenvironment
EMTepithelial-mesenchymal transition
CAFcancer-associated fibroblast
TAMtumor-associated macrophage
PFSprogression-free survival
EOMESeomesodermin
TCGAThe Cancer Genome Atlas
OSoverall survival
ORRoverall response rate
DCRdisease control rate
IRAEimmune-related adverse event

References

  1. Siegel, R.L.; Miller, K.D.; Wagle, N.S.; Jemal, A. Cancer statistics, 2023. CA Cancer J. Clin. 2023, 73, 17–48. [Google Scholar] [CrossRef] [PubMed]
  2. Perez-Moreno, P.; Brambilla, E.; Thomas, R.; Soria, J.-C. Squamous Cell Carcinoma of the Lung: Molecular Subtypes and Therapeutic Opportunities. Clin. Cancer Res. 2012, 18, 2443–2451. [Google Scholar] [CrossRef] [PubMed]
  3. Thai, A.A.; Solomon, B.J.; Sequist, L.V.; Gainor, J.F.; Heist, R.S. Lung cancer. Lancet 2021, 398, 535–554. [Google Scholar] [CrossRef]
  4. Midha, A.; Dearden, S.; McCormack, R. EGFR mutation incidence in non-small-cell lung cancer of adenocarcinoma histology: A systematic review and global map by ethnicity (mutMapII). Am. J. Cancer Res. 2015, 5, 2892–2911. [Google Scholar]
  5. Gou, Q.; Gou, Q.; Gan, X.; Xie, Y. Novel therapeutic strategies for rare mutations in non-small cell lung cancer. Sci. Rep. 2024, 14, 10317. [Google Scholar] [CrossRef]
  6. Tsuboi, M.; Herbst, R.S.; John, T.; Kato, T.; Majem, M.; Grohe, C.; Wang, J.; Goldman, J.W.; Lu, S.; Su, W.C.; et al. Overall Survival with Osimertinib in Resected EGFR-Mutated NSCLC. N. Engl. J. Med. 2023, 389, 137–147. [Google Scholar] [CrossRef] [PubMed]
  7. Planchard, D.; Janne, P.A.; Cheng, Y.; Yang, J.C.; Yanagitani, N.; Kim, S.W.; Sugawara, S.; Yu, Y.; Fan, Y.; Geater, S.L.; et al. Osimertinib with or without Chemotherapy in EGFR-Mutated Advanced NSCLC. N. Engl. J. Med. 2023, 389, 1935–1948. [Google Scholar] [CrossRef]
  8. Cho, B.C.; Lu, S.; Felip, E.; Spira, A.I.; Girard, N.; Lee, J.S.; Lee, S.H.; Ostapenko, Y.; Danchaivijitr, P.; Liu, B.; et al. Amivantamab plus Lazertinib in Previously Untreated EGFR-Mutated Advanced NSCLC. N. Engl. J. Med. 2024, 391, 1486–1498. [Google Scholar] [CrossRef]
  9. Koulouris, A.; Tsagkaris, C.; Corriero, A.C.; Metro, G.; Mountzios, G. Resistance to TKIs in EGFR-Mutated Non-Small Cell Lung Cancer: From Mechanisms to New Therapeutic Strategies. Cancers 2022, 14, 3337. [Google Scholar] [CrossRef]
  10. Reita, D.; Pabst, L.; Pencreach, E.; Guérin, E.; Dano, L.; Rimelen, V.; Voegeli, A.-C.; Vallat, L.; Mascaux, C.; Beau-Faller, M. Molecular Mechanism of EGFR-TKI Resistance in EGFR-Mutated Non-Small Cell Lung Cancer: Application to Biological Diagnostic and Monitoring. Cancers 2021, 13, 4926. [Google Scholar] [CrossRef]
  11. Sekine, Y.; Hata, A.; Koh, E.; Hiroshima, K. Lung carcinogenesis from chronic obstructive pulmonary disease: Characteristics of lung cancer from COPD and contribution of signal transducers and lung stem cells in the inflammatory microenvironment. Gen. Thorac. Cardiovasc. Surg. 2014, 62, 415–421. [Google Scholar] [CrossRef]
  12. Tan, Z.; Xue, H.; Sun, Y.; Zhang, C.; Song, Y.; Qi, Y. The Role of Tumor Inflammatory Microenvironment in Lung Cancer. Front. Pharmacol. 2021, 12, 688625. [Google Scholar] [CrossRef]
  13. Heinrich, E.L.; Walser, T.C.; Krysan, K.; Liclican, E.L.; Grant, J.L.; Rodriguez, N.L.; Dubinett, S.M. The inflammatory tumor microenvironment, epithelial mesenchymal transition and lung carcinogenesis. Cancer Microenviron. 2012, 5, 5–18. [Google Scholar] [CrossRef]
  14. Yuan, S.; Almagro, J.; Fuchs, E. Beyond genetics: Driving cancer with the tumour microenvironment behind the wheel. Nat. Rev. Cancer 2024, 24, 274–286. [Google Scholar] [CrossRef]
  15. Cacho-Diaz, B.; Garcia-Botello, D.R.; Wegman-Ostrosky, T.; Reyes-Soto, G.; Ortiz-Sanchez, E.; Herrera-Montalvo, L.A. Tumor microenvironment differences between primary tumor and brain metastases. J. Transl. Med. 2020, 18, 1. [Google Scholar] [CrossRef]
  16. Kundu, M.; Butti, R.; Panda, V.K.; Malhotra, D.; Das, S.; Mitra, T.; Kapse, P.; Gosavi, S.W.; Kundu, G.C. Modulation of the tumor microenvironment and mechanism of immunotherapy-based drug resistance in breast cancer. Mol. Cancer 2024, 23, 92. [Google Scholar] [CrossRef]
  17. Falcone, I.; Conciatori, F.; Bazzichetto, C.; Ferretti, G.; Cognetti, F.; Ciuffreda, L.; Milella, M. Tumor Microenvironment: Implications in Melanoma Resistance to Targeted Therapy and Immunotherapy. Cancers 2020, 12, 2870. [Google Scholar] [CrossRef]
  18. Madeddu, C.; Donisi, C.; Liscia, N.; Lai, E.; Scartozzi, M.; Maccio, A. EGFR-Mutated Non-Small Cell Lung Cancer and Resistance to Immunotherapy: Role of the Tumor Microenvironment. Int. J. Mol. Sci. 2022, 23, 6489. [Google Scholar] [CrossRef]
  19. Maemondo, M.; Inoue, A.; Kobayashi, K.; Sugawara, S.; Oizumi, S.; Isobe, H.; Gemma, A.; Harada, M.; Yoshizawa, H.; Kinoshita, I.; et al. Gefitinib or Chemotherapy for Non-Small-Cell Lung Cancer with Mutated EGFR. N. Engl. J. Med. 2010, 362, 2380–2388. [Google Scholar] [CrossRef]
  20. Cataldo, V.D.; Gibbons, D.L.; Pérez-Soler, R.; Quintás-Cardama, A. Treatment of Non-Small-Cell Lung Cancer with Erlotinib or Gefitinib. N. Engl. J. Med. 2011, 364, 947–955. [Google Scholar] [CrossRef]
  21. Soria, J.-C.; Felip, E.; Cobo, M.; Lu, S.; Syrigos, K.; Lee, K.H.; Göker, E.; Georgoulias, V.; Li, W.; Isla, D.; et al. Afatinib versus erlotinib as second-line treatment of patients with advanced squamous cell carcinoma of the lung (LUX-Lung 8): An open-label randomised controlled phase 3 trial. Lancet Oncol. 2015, 16, 897–907. [Google Scholar] [CrossRef]
  22. Soria, J.-C.; Ohe, Y.; Vansteenkiste, J.; Reungwetwattana, T.; Chewaskulyong, B.; Lee, K.H.; Dechaphunkul, A.; Imamura, F.; Nogami, N.; Kurata, T.; et al. Osimertinib in Untreated EGFR-Mutated Advanced Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 378, 113–125. [Google Scholar] [CrossRef]
  23. Cho, B.C.; Ahn, M.-J.; Kang, J.H.; Soo, R.A.; Reungwetwattana, T.; Yang, J.C.-H.; Cicin, I.; Kim, D.-W.; Wu, Y.-L.; Lu, S.; et al. Lazertinib Versus Gefitinib as First-Line Treatment in Patients with EGFR-Mutated Advanced Non–Small-Cell Lung Cancer: Results From LASER301. J. Clin. Oncol. 2023, 41, 4208–4217. [Google Scholar] [CrossRef]
  24. Mok, T.S.; Cheng, Y.; Zhou, X.; Lee, K.H.; Nakagawa, K.; Niho, S.; Lee, M.; Linke, R.; Rosell, R.; Corral, J.; et al. Improvement in Overall Survival in a Randomized Study That Compared Dacomitinib with Gefitinib in Patients with Advanced Non-Small-Cell Lung Cancer and EGFR-Activating Mutations. J. Clin. Oncol. 2018, 36, 2244–2250. [Google Scholar] [CrossRef]
  25. 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 Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2017, 377, 829–838. [Google Scholar] [CrossRef]
  26. Shaw, A.T.; Kim, D.-W.; Mehra, R.; Tan, D.S.W.; Felip, E.; Chow, L.Q.M.; Camidge, D.R.; Vansteenkiste, J.; Sharma, S.; Pas, T.D.; et al. Ceritinib in ALK-Rearranged Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2014, 370, 1189–1197. [Google Scholar] [CrossRef]
  27. Camidge, D.R.; Kim, H.R.; Ahn, M.-J.; Yang, J.C.-H.; Han, J.-Y.; Lee, J.-S.; Hochmair, M.J.; Li, J.Y.-C.; Chang, G.-C.; Lee, K.H.; et al. Brigatinib versus Crizotinib in ALK-Positive Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 379, 2027–2039. [Google Scholar] [CrossRef]
  28. Shaw, A.T.; Bauer, T.M.; Marinis, F.d.; 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]
  29. Shaw, A.T.; Kim, D.-W.; Nakagawa, K.; Seto, T.; Crinó, L.; Ahn, M.-J.; Pas, T.D.; Besse, B.; Solomon, B.J.; Blackhall, F.; et al. Crizotinib versus Chemotherapy in Advanced ALK-Positive Lung Cancer. N. Engl. J. Med. 2013, 368, 2385–2394. [Google Scholar] [CrossRef]
  30. Wolf, J.; Seto, T.; Han, J.-Y.; Reguart, N.; Garon, E.B.; Groen, H.J.M.; Tan, D.S.W.; Hida, T.; Jonge, M.d.; Orlov, S.V.; et al. Capmatinib in MET Exon 14–Mutated or MET-Amplified Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2020, 383, 944–957. [Google Scholar] [CrossRef]
  31. Paik, P.K.; Felip, E.; Veillon, R.; Sakai, H.; Cortot, A.B.; Garassino, M.C.; Mazieres, J.; Viteri, S.; Senellart, H.; Meerbeeck, J.V.; et al. Tepotinib in Non-Small-Cell Lung Cancer with MET Exon 14 Skipping Mutations. N. Engl. J. Med. 2020, 383, 931–943. [Google Scholar] [CrossRef]
  32. 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 Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2020, 383, 813–824. [Google Scholar] [CrossRef]
  33. 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]
  34. Zhou, C.; Tang, K.-J.; Cho, B.C.; Liu, B.; Paz-Ares, L.; Cheng, S.; Kitazono, S.; Thiagarajan, M.; Goldman, J.W.; Sabari, J.K.; et al. Amivantamab plus Chemotherapy in NSCLC with EGFR Exon 20 Insertions. N. Engl. J. Med. 2023, 389, 2039–2051. [Google Scholar] [CrossRef]
  35. 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 Platinum-Pretreated Patients with EGFR Exon 20 Insertion–Positive Metastatic Non–Small Cell Lung Cancer: A Phase 1/2 Open-label Nonrandomized Clinical Trial. JAMA Oncol. 2021, 7, e214761. [Google Scholar] [CrossRef]
  36. Planchard, D.; Besse, B.; Groen, H.J.M.; Hashemi, S.M.S.; Mazieres, J.; Kim, T.M.; Quoix, E.; Souquet, P.-J.; Barlesi, F.; Baik, C.; et al. Phase 2 Study of Dabrafenib Plus Trametinib in Patients with BRAF V600E-Mutant Metastatic NSCLC: Updated 5-Year Survival Rates and Genomic Analysis. J. Thorac. Oncol. 2022, 17, 103–115. [Google Scholar] [CrossRef]
  37. Stinchcombe, T.E. Encorafenib and Binimetinib: A New Treatment Option for BRAFV600E-Mutant Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2023, 41, 3679–3681. [Google Scholar] [CrossRef]
  38. Purba, E.R.; Saita, E.I.; Maruyama, I.N. Activation of the EGF Receptor by Ligand Binding and Oncogenic Mutations: The “Rotation Model”. Cells 2017, 6, 13. [Google Scholar] [CrossRef]
  39. Rosell, R.; Cardona, A.F.; Arrieta, O.; Aguilar, A.; Ito, M.; Pedraz, C.; Codony-Servat, J.; Santarpia, M. Coregulation of pathways in lung cancer patients with EGFR mutation: Therapeutic opportunities. Br. J. Cancer 2021, 125, 1602–1611. [Google Scholar] [CrossRef]
  40. Kobayashi, K.; Tan, A.C. Unraveling the Impact of Intratumoral Heterogeneity on EGFR Tyrosine Kinase Inhibitor Resistance in EGFR-Mutated NSCLC. Int. J. Mol. Sci. 2023, 24, 4126. [Google Scholar] [CrossRef]
  41. Meador, C.B.; Hata, A.N. Acquired resistance to targeted therapies in NSCLC: Updates and evolving insights. Pharmacol. Ther. 2020, 210, 107522. [Google Scholar] [CrossRef]
  42. Chhouri, H.; Alexandre, D.; Grumolato, L. Mechanisms of Acquired Resistance and Tolerance to EGFR Targeted Therapy in Non-Small Cell Lung Cancer. Cancers 2023, 15, 504. [Google Scholar] [CrossRef]
  43. Li, Y.; Mao, T.; Wang, J.; Zheng, H.; Hu, Z.; Cao, P.; Yang, S.; Zhu, L.; Guo, S.; Zhao, X.; et al. Toward the next generation EGFR inhibitors: An overview of osimertinib resistance mediated by EGFR mutations in non-small cell lung cancer. Cell Commun. Signal 2023, 21, 71. [Google Scholar] [CrossRef]
  44. Takezawa, K.; Pirazzoli, V.; Arcila, M.E.; Nebhan, C.A.; Song, X.; de Stanchina, E.; Ohashi, K.; Janjigian, Y.Y.; Spitzler, P.J.; Melnick, M.A. HER2 amplification: A potential mechanism of acquired resistance to EGFR inhibition in EGFR-mutant lung cancers that lack the second-site EGFR T790M mutation. Cancer Discov. 2012, 2, 922–933. [Google Scholar] [CrossRef]
  45. Yamaoka, T.; Ohmori, T.; Ohba, M.; Arata, S.; Murata, Y.; Kusumoto, S.; Ando, K.; Ishida, H.; Ohnishi, T.; Sasaki, Y. Distinct Afatinib Resistance Mechanisms Identified in Lung Adenocarcinoma Harboring an EGFR Mutation. Mol. Cancer Res. 2017, 15, 915–928. [Google Scholar] [CrossRef] [PubMed]
  46. Rotow, J.K.; Lee, J.K.; Madison, R.W.; Oxnard, G.R.; Janne, P.A.; Schrock, A.B. Real-World Genomic Profile of EGFR Second-Site Mutations and Other Osimertinib Resistance Mechanisms and Clinical Landscape of NSCLC Post-Osimertinib. J. Thorac. Oncol. 2024, 19, 227–239. [Google Scholar] [CrossRef]
  47. Gomatou, G.; Syrigos, N.; Kotteas, E. Osimertinib Resistance: Molecular Mechanisms and Emerging Treatment Options. Cancers 2023, 15, 841. [Google Scholar] [CrossRef]
  48. Niederst, M.J.; Engelman, J.A. Bypass mechanisms of resistance to receptor tyrosine kinase inhibition in lung cancer. Sci. Signal 2013, 6, re6. [Google Scholar] [CrossRef]
  49. Chmielecki, J.; Gray, J.E.; Cheng, Y.; Ohe, Y.; Imamura, F.; Cho, B.C.; Lin, M.C.; Majem, M.; Shah, R.; Rukazenkov, Y.; et al. Candidate mechanisms of acquired resistance to first-line osimertinib in EGFR-mutated advanced non-small cell lung cancer. Nat. Commun. 2023, 14, 1070. [Google Scholar] [CrossRef] [PubMed]
  50. Fassunke, J.; Muller, F.; Keul, M.; Michels, S.; Dammert, M.A.; Schmitt, A.; Plenker, D.; Lategahn, J.; Heydt, C.; Bragelmann, J.; et al. Overcoming EGFR(G724S)-mediated osimertinib resistance through unique binding characteristics of second-generation EGFR inhibitors. Nat. Commun. 2018, 9, 4655. [Google Scholar] [CrossRef]
  51. Lim, S.M.; Cho, B.C.; Han, J.-Y.; Kim, S.-W.; Lee, K.H.; Nagasaka, M.; Jo, A.; Seah, E.; Kim, C.; Reungwetwattana, T. Phase 1/2 clinical trial of JIN-A02, a 4th generation EGFR-TKI, in patients with 3rd generation EGFR-TKI resistance in EGFR mutated advanced/metastatic non-small cell lung cancer (NSCLC). J. Clin. Oncol. 2024, 42, TPS8658. [Google Scholar] [CrossRef]
  52. Phase 1/2 Study of BDTX-1535 in Patients with Glioblastoma or Non-Small Cell Lung Cancer with EGFR Mutations. Available online: https://clinicaltrials.gov/study/NCT05256290 (accessed on 23 September 2024).
  53. Engelman, J.A.; Zejnullahu, K.; Mitsudomi, T.; Song, Y.; Hyland, C.; Park, J.O.; Lindeman, N.; Gale, C.M.; Zhao, X.; Christensen, J.; et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 2007, 316, 1039–1043. [Google Scholar] [CrossRef] [PubMed]
  54. Bean, J.; Brennan, C.; Shih, J.Y.; Riely, G.; Viale, A.; Wang, L.; Chitale, D.; Motoi, N.; Szoke, J.; Broderick, S.; et al. MET amplification occurs with or without T790M mutations in EGFR mutant lung tumors with acquired resistance to gefitinib or erlotinib. Proc. Natl. Acad. Sci. USA 2007, 104, 20932–20937. [Google Scholar] [CrossRef]
  55. Papadimitrakopoulou, V.A.; Wu, Y.L.; Han, J.Y.; Ahn, M.J.; Ramalingam, S.S.; John, T.; Okamoto, I.; Yang, J.C.H.; Bulusu, K.C.; Laus, G.; et al. Analysis of resistance mechanisms to osimertinib in patients with EGFR T790M advanced NSCLC from the AURA3 study. Ann. Oncol. 2018, 29, viii741. [Google Scholar] [CrossRef]
  56. Coleman, N.; Hong, L.; Zhang, J.; Heymach, J.; Hong, D.; Le, X. Beyond epidermal growth factor receptor: MET amplification as a general resistance driver to targeted therapy in oncogene-driven non-small-cell lung cancer. ESMO Open 2021, 6, 100319. [Google Scholar] [CrossRef] [PubMed]
  57. Wang, Y.; Li, L.; Han, R.; Jiao, L.; Zheng, J.; He, Y. Clinical analysis by next-generation sequencing for NSCLC patients with MET amplification resistant to osimertinib. Lung Cancer 2018, 118, 105–110. [Google Scholar] [CrossRef]
  58. Zhu, V.W.; Schrock, A.B.; Ali, S.M.; Ou, S.I. Differential response to a combination of full-dose osimertinib and crizotinib in a patient with EGFR-mutant non-small cell lung cancer and emergent MET amplification. Lung Cancer 2019, 10, 21–26. [Google Scholar] [CrossRef] [PubMed]
  59. Smit, E.F.; Dooms, C.; Raskin, J.; Nadal, E.; Tho, L.M.; Le, X.; Mazieres, J.; Hin, H.S.; Morise, M.; Zhu, V.W.; et al. INSIGHT 2: A phase II study of tepotinib plus osimertinib in MET-amplified NSCLC and first-line osimertinib resistance. Future Oncol. 2022, 18, 1039–1054. [Google Scholar] [CrossRef] [PubMed]
  60. Ahn, M.j.; De Marinis, F.; Bonanno, L.; Cho, B.C.; Kim, T.M.; Cheng, S.; Novello, S.; Proto, C.; Kim, S.W.; Lee, J.S.; et al. EP08.02-140 MET Biomarker-based Preliminary Efficacy Analysis in SAVANNAH: Savolitinib+osimertinib in EGFRm NSCLC Post-Osimertinib. J. Thorac. Oncol. 2022, 17, S469–S470. [Google Scholar] [CrossRef]
  61. Kim, T.M.; Guarneri, V.; Jye, V.P.; Khaw, L.B.; Yang, J.J.; Wislez, M.; Huang, C.; Chong Kin, L.; Mazieres, J.; Tho, L.M.; et al. OA21.05 Tepotinib + Osimertinib in EGFR-mutant NSCLC with MET Amplification Following 1L Osimertinib: INSIGHT 2 Primary Analysis. J. Thorac. Oncol. 2023, 18, S94. [Google Scholar] [CrossRef]
  62. Yu, H.A.; Ambrose, H.; Baik, C.; Cho, B.C.; Cocco, E.; Goldberg, S.B.; Goldman, J.W.; Kraljevic, S.; de Langen, A.J.; Okamoto, I.; et al. 1239P ORCHARD osimertinib + savolitinib interim analysis: A biomarker-directed phase II platform study in patients (pts) with advanced non-small cell lung cancer (NSCLC) whose disease has progressed on first-line (1L) osimertinib. Ann. Oncol. 2021, 32, S978–S979. [Google Scholar] [CrossRef]
  63. Liam, C.K.; Ahmad, A.R.; Hsia, T.C.; Zhou, J.; Kim, D.W.; Soo, R.A.; Cheng, Y.; Lu, S.; Shin, S.W.; Yang, J.C.; et al. Randomized Trial of Tepotinib Plus Gefitinib versus Chemotherapy in EGFR-Mutant NSCLC with EGFR Inhibitor Resistance Due to MET Amplification: INSIGHT Final Analysis. Clin. Cancer Res. 2023, 29, 1879–1886. [Google Scholar] [CrossRef] [PubMed]
  64. Besse, B.; Baik, C.S.; Marmarelis, M.E.; Sabari, J.K.; Goto, K.; Shu, C.A.; Lee, J.-S.; Ou, S.-H.I.; Cho, B.C.; Waqar, S.N.; et al. Predictive biomarkers for treatment with amivantamab plus lazertinib among EGFR-mutated NSCLC in the post-osimertinib setting: Analysis of tissue IHC and ctDNA NGS. J. Clin. Oncol. 2023, 41, 9013. [Google Scholar] [CrossRef]
  65. Wu, Y.L.; Guarneri, V.; Voon, P.J.; Lim, B.K.; Yang, J.J.; Wislez, M.; Huang, C.; Liam, C.K.; Mazieres, J.; Tho, L.M.; et al. Tepotinib plus osimertinib in patients with EGFR-mutated non-small-cell lung cancer with MET amplification following progression on first-line osimertinib (INSIGHT 2): A multicentre, open-label, phase 2 trial. Lancet Oncol. 2024, 25, 989–1002. [Google Scholar] [CrossRef]
  66. Bertoli, E.; De Carlo, E.; Del Conte, A.; Stanzione, B.; Revelant, A.; Fassetta, K.; Spina, M.; Bearz, A. Acquired Resistance to Osimertinib in EGFR-Mutated Non-Small Cell Lung Cancer: How Do We Overcome It? Int. J. Mol. Sci. 2022, 23, 6936. [Google Scholar] [CrossRef] [PubMed]
  67. Chmielecki, J.; Mok, T.; Wu, Y.L.; Han, J.Y.; Ahn, M.J.; Ramalingam, S.S.; John, T.; Okamoto, I.; Yang, J.C.; Shepherd, F.A.; et al. Analysis of acquired resistance mechanisms to osimertinib in patients with EGFR-mutated advanced non-small cell lung cancer from the AURA3 trial. Nat. Commun. 2023, 14, 1071. [Google Scholar] [CrossRef] [PubMed]
  68. Ramalingam, S.S.; Cheng, Y.; Zhou, C.; Ohe, Y.; Imamura, F.; Cho, B.C.; Lin, M.C.; Majem, M.; Shah, R.; Rukazenkov, Y.; et al. Mechanisms of acquired resistance to first-line osimertinib: Preliminary data from the phase III FLAURA study. Ann. Oncol. 2018, 29, viii740. [Google Scholar] [CrossRef]
  69. Lee, K.; Jung, H.A.; Sun, J.M.; Lee, S.H.; Ahn, J.S.; Park, K.; Ahn, M.J. Clinical Characteristics and Outcomes of Non-small Cell Lung Cancer Patients with HER2 Alterations in Korea. Cancer Res. Treat. 2020, 52, 292–300. [Google Scholar] [CrossRef]
  70. Yoshizawa, A.; Sumiyoshi, S.; Sonobe, M.; Kobayashi, M.; Uehara, T.; Fujimoto, M.; Tsuruyama, T.; Date, H.; Haga, H. HER2 status in lung adenocarcinoma: A comparison of immunohistochemistry, fluorescence in situ hybridization (FISH), dual-ISH, and gene mutations. Lung Cancer 2014, 85, 373–378. [Google Scholar] [CrossRef] [PubMed]
  71. Mazieres, J.; Barlesi, F.; Filleron, T.; Besse, B.; Monnet, I.; Beau-Faller, M.; Peters, S.; Dansin, E.; Fruh, M.; Pless, M.; et al. Lung cancer patients with HER2 mutations treated with chemotherapy and HER2-targeted drugs: Results from the European EUHER2 cohort. Ann. Oncol. 2016, 27, 281–286. [Google Scholar] [CrossRef] [PubMed]
  72. Ferrari, G.; Del Rio, B.; Novello, S.; Passiglia, F. HER2-Altered Non-Small Cell Lung Cancer: A Journey from Current Approaches to Emerging Strategies. Cancers 2024, 16, 2018. [Google Scholar] [CrossRef]
  73. Jebbink, M.; de Langen, A.J.; Monkhorst, K.; Boelens, M.C.; van den Broek, D.; van der Noort, V.; de Gooijer, C.J.; Mahn, M.; van der Wekken, A.J.; Hendriks, L.; et al. Trastuzumab-Emtansine and Osimertinib Combination Therapy to Target HER2 Bypass Track Resistance in EGFR Mutation-Positive NSCLC. JTO Clin. Res. Rep. 2023, 4, 100481. [Google Scholar] [CrossRef] [PubMed]
  74. Romaniello, D.; Marrocco, I.; Belugali Nataraj, N.; Ferrer, I.; Drago-Garcia, D.; Vaknin, I.; Oren, R.; Lindzen, M.; Ghosh, S.; Kreitman, M.; et al. Targeting HER3, a Catalytically Defective Receptor Tyrosine Kinase, Prevents Resistance of Lung Cancer to a Third-Generation EGFR Kinase Inhibitor. Cancers 2020, 12, 2394. [Google Scholar] [CrossRef] [PubMed]
  75. Chen, Q.; Jia, G.; Zhang, X.; Ma, W. Targeting HER3 to overcome EGFR TKI resistance in NSCLC. Front. Immunol. 2023, 14, 1332057. [Google Scholar] [CrossRef] [PubMed]
  76. Fu, Z.; Li, S.; Han, S.; Shi, C.; Zhang, Y. Antibody drug conjugate: The “biological missile” for targeted cancer therapy. Signal Transduct. Target. Ther. 2022, 7, 93. [Google Scholar] [CrossRef]
  77. Yu, H.A.; Baik, C.; Kim, D.W.; Johnson, M.L.; Hayashi, H.; Nishio, M.; Yang, J.C.; Su, W.C.; Gold, K.A.; Koczywas, M.; et al. Translational insights and overall survival in the U31402-A-U102 study of patritumab deruxtecan (HER3-DXd) in EGFR-mutated NSCLC. Ann. Oncol. 2024, 35, 437–447. [Google Scholar] [CrossRef]
  78. Zhang, L.; Ma, Y.; Zhao, Y.; Fang, W.F.; Zhao, H.; Huang, Y.; Yang, Y.; Hou, X.; Wen, Z.; Zhang, S.; et al. 1316MO BL-B01D1, a first-in-class EGFRxHER3 bispecific antibody-drug conjugate, in patients with non-small cell lung cancer: Updated results from first-in-human phase I study. Ann. Oncol. 2023, 34, S758. [Google Scholar] [CrossRef]
  79. Paz-Ares, L.; Ahn, M.J.; Lisberg, A.E.; Kitazono, S.; Cho, B.C.; Blumenschein, G.; Shum, E.; Pons-Tostivint, E.; Goto, Y.; Yoh, K.; et al. 1314MO TROPION-Lung05: Datopotamab deruxtecan (Dato-DXd) in previously treated non-small cell lung cancer (NSCLC) with actionable genomic alterations (AGAs). Ann. Oncol. 2023, 34, S755–S756. [Google Scholar] [CrossRef]
  80. Horinouchi, H.; Cho, B.C.; Camidge, D.R.; Goto, K.; Tomasini, P.; Li, Y.; Vasilopoulos, A.; Brunsdon, P.; Hoffman, D.; Shi, W.; et al. 515MO Phase Ib study of telisotuzumab vedotin (Teliso-V) and osimertinib in patients (Pts) with advanced EGFR-mutated (Mut), c-Met overexpressing (OE) non-small cell lung cancer (NSCLC): Final efficacy and safety updates. Ann. Oncol. 2023, 34, S1670. [Google Scholar] [CrossRef]
  81. Mok, T.; Janne, P.A.; Nishio, M.; Novello, S.; Reck, M.; Steuer, C.; Wu, Y.L.; Fougeray, R.; Fan, P.D.; Meng, J.; et al. HERTHENA-Lung02: Phase III study of patritumab deruxtecan in advanced EGFR-mutated NSCLC after a third-generation EGFR TKI. Future Oncol. 2024, 20, 969–980. [Google Scholar] [CrossRef] [PubMed]
  82. Bing, Z.; Wang, W.; Wang, D.; Ma, T. Identification of RET fusion as mechanisms of resistance to EGFR tyrosine-kinase inhibitors. J. Clin. Oncol. 2021, 39, e21074. [Google Scholar] [CrossRef]
  83. Yao, Y.; Zhang, M.; Liu, X.; Zhao, J.; Cheng, X.; Zeng, A.; Kong, J.; Zhang, H.; Chen, R.; Xia, X. RET fusion in first/third-generation EGFR-TKIs resistance in advanced non-small cell lung cancer. J. Clin. Oncol. 2019, 37, e20634. [Google Scholar] [CrossRef]
  84. Wang, C.; Zhang, Z.; Sun, Y.; Wang, S.; Wu, M.; Ou, Q.; Xu, Y.; Chen, Z.; Shao, Y.; Liu, H.; et al. RET fusions as primary oncogenic drivers and secondary acquired resistance to EGFR tyrosine kinase inhibitors in patients with non-small-cell lung cancer. J. Transl. Med. 2022, 20, 390. [Google Scholar] [CrossRef] [PubMed]
  85. Rotow, J.; Patel, J.D.; Hanley, M.P.; Yu, H.; Awad, M.; Goldman, J.W.; Nechushtan, H.; Scheffler, M.; Kuo, C.S.; Rajappa, S.; et al. Osimertinib and Selpercatinib Efficacy, Safety, and Resistance in a Multicenter, Prospectively Treated Cohort of EGFR-Mutant and RET Fusion-Positive Lung Cancers. Clin. Cancer Res. 2023, 29, 2979–2987. [Google Scholar] [CrossRef] [PubMed]
  86. Xie, Z.; Gu, Y.; Xie, X.; Lin, X.; Ouyang, M.; Qin, Y.; Zhang, J.; Lizaso, A.; Chen, S.; Zhou, C. Lung Adenocarcinoma Harboring Concomitant EGFR Mutations and BRAF V600E Responds to a Combination of Osimertinib and Vemurafenib to Overcome Osimertinib Resistance. Clin. Lung Cancer 2021, 22, e390–e394. [Google Scholar] [CrossRef]
  87. Meng, P.; Koopman, B.; Kok, K.; Ter Elst, A.; Schuuring, E.; van Kempen, L.C.; Timens, W.; Hiltermann, T.J.N.; Groen, H.J.M.; van den Berg, A.; et al. Combined osimertinib, dabrafenib and trametinib treatment for advanced non-small-cell lung cancer patients with an osimertinib-induced BRAF V600E mutation. Lung Cancer 2020, 146, 358–361. [Google Scholar] [CrossRef]
  88. Iksen; Pothongsrisit, S.; Pongrakhananon, V. Targeting the PI3K/AKT/mTOR Signaling Pathway in Lung Cancer: An Update Regarding Potential Drugs and Natural Products. Molecules 2021, 26, 4100. [Google Scholar] [CrossRef] [PubMed]
  89. Liu, X.; Mei, W.; Zhang, P.; Zeng, C. PIK3CA mutation as an acquired resistance driver to EGFR-TKIs in non-small cell lung cancer: Clinical challenges and opportunities. Pharmacol. Res. 2024, 202, 107123. [Google Scholar] [CrossRef]
  90. Ramachandran, S.; Verma, A.K.; Dev, K.; Goyal, Y.; Bhatt, D.; Alsahli, M.A.; Rahmani, A.H.; Almatroudi, A.; Almatroodi, S.A.; Alrumaihi, F.; et al. Role of Cytokines and Chemokines in NSCLC Immune Navigation and Proliferation. Oxidative Med. Cell Longev. 2021, 2021, 5563746. [Google Scholar] [CrossRef] [PubMed]
  91. Hanahan, D.; Coussens, L.M. Accessories to the crime: Functions of cells recruited to the tumor microenvironment. Cancer Cell 2012, 21, 309–322. [Google Scholar] [CrossRef]
  92. Chen, S.; Tang, J.; Liu, F.; Li, W.; Yan, T.; Shangguan, D.; Yang, N.; Liao, D. Changes of tumor microenvironment in non-small cell lung cancer after TKI treatments. Front. Immunol. 2023, 14, 1094764. [Google Scholar] [CrossRef]
  93. Ding, H.; Wu, L.; Qin, H.; Fu, W.; Wang, Y.; Wu, M.; Wang, J.; Han, Y. Synergistic Anti-Tumor Efficacy Achieved by Reversing Drug Resistance through the Regulation of the Tumor Immune Microenvironment with IL-12 and Osimertinib Combination Therapy. J. Cancer 2024, 15, 4534–4550. [Google Scholar] [CrossRef]
  94. Xiang, X.; Wang, J.; Lu, D.; Xu, X. Targeting tumor-associated macrophages to synergize tumor immunotherapy. Signal Transduct. Target. Ther. 2021, 6, 75. [Google Scholar] [CrossRef] [PubMed]
  95. Lim, J.U.; Lee, E.; Lee, S.Y.; Cho, H.J.; Ahn, D.H.; Hwang, Y.; Choi, J.Y.; Yeo, C.D.; Park, C.K.; Kim, S.J. Current literature review on the tumor immune micro-environment, its heterogeneity and future perspectives in treatment of advanced non-small cell lung cancer. Transl. Lung Cancer Res. 2023, 12, 857–876. [Google Scholar] [CrossRef] [PubMed]
  96. Li, L.; Lu, G.; Liu, Y.; Gong, L.; Zheng, X.; Zheng, H.; Gu, W.; Yang, L. Low Infiltration of CD8+ PD-L1+ T Cells and M2 Macrophages Predicts Improved Clinical Outcomes After Immune Checkpoint Inhibitor Therapy in Non-Small Cell Lung Carcinoma. Front. Oncol. 2021, 11, 658690. [Google Scholar] [CrossRef] [PubMed]
  97. Fang, Y.; Wang, Y.; Zeng, D.; Zhi, S.; Shu, T.; Huang, N.; Zheng, S.; Wu, J.; Liu, Y.; Huang, G.; et al. Comprehensive analyses reveal TKI-induced remodeling of the tumor immune microenvironment in EGFR/ALK-positive non-small-cell lung cancer. Oncoimmunology 2021, 10, 1951019. [Google Scholar] [CrossRef] [PubMed]
  98. Isomoto, K.; Haratani, K.; Hayashi, H.; Shimizu, S.; Tomida, S.; Niwa, T.; Yokoyama, T.; Fukuda, Y.; Chiba, Y.; Kato, R.; et al. Impact of EGFR-TKI Treatment on the Tumor Immune Microenvironment in EGFR Mutation-Positive Non-Small Cell Lung Cancer. Clin. Cancer Res. 2020, 26, 2037–2046. [Google Scholar] [CrossRef]
  99. Kawana, S.; Saito, R.; Miki, Y.; Kimura, Y.; Abe, J.; Sato, I.; Endo, M.; Sugawara, S.; Sasano, H. Suppression of tumor immune microenvironment via microRNA-1 after epidermal growth factor receptor-tyrosine kinase inhibitor resistance acquirement in lung adenocarcinoma. Cancer Med. 2021, 10, 718–727. [Google Scholar] [CrossRef]
  100. Maynard, A.; McCoach, C.E.; Rotow, J.K.; Harris, L.; Haderk, F.; Kerr, D.L.; Yu, E.A.; Schenk, E.L.; Tan, W.; Zee, A.; et al. Therapy-Induced Evolution of Human Lung Cancer Revealed by Single-Cell RNA Sequencing. Cell 2020, 182, 1232–1251.e1222. [Google Scholar] [CrossRef] [PubMed]
  101. Miller, C.H.; Klawon, D.E.J.; Zeng, S.; Lee, V.; Socci, N.D.; Savage, P.A. Eomes identifies thymic precursors of self-specific memory-phenotype CD8(+) T cells. Nat. Immunol. 2020, 21, 567–577. [Google Scholar] [CrossRef] [PubMed]
  102. van der Woude, L.L.; Gorris, M.A.J.; Halilovic, A.; Figdor, C.G.; de Vries, I.J.M. Migrating into the Tumor: A Roadmap for T Cells. Trends Cancer 2017, 3, 797–808. [Google Scholar] [CrossRef]
  103. Han, R.; Guo, H.; Shi, J.; Wang, H.; Zhao, S.; Jia, Y.; Liu, X.; Li, J.; Cheng, L.; Zhao, C.; et al. Tumour microenvironment changes after osimertinib treatment resistance in non-small cell lung cancer. Eur. J. Cancer 2023, 189, 112919. [Google Scholar] [CrossRef] [PubMed]
  104. Zhu, X.; Zhu, J. CD4 T Helper Cell Subsets and Related Human Immunological Disorders. Int. J. Mol. Sci. 2020, 21, 8011. [Google Scholar] [CrossRef]
  105. Li, C.; Jiang, P.; Wei, S.; Xu, X.; Wang, J. Regulatory T cells in tumor microenvironment: New mechanisms, potential therapeutic strategies and future prospects. Mol. Cancer 2020, 19, 116. [Google Scholar] [CrossRef]
  106. Sugiyama, E.; Togashi, Y.; Takeuchi, Y.; Shinya, S.; Tada, Y.; Kataoka, K.; Tane, K.; Sato, E.; Ishii, G.; Goto, K.; et al. Blockade of EGFR improves responsiveness to PD-1 blockade in EGFR-mutated non-small cell lung cancer. Sci. Immunol. 2020, 5, eaav3937. [Google Scholar] [CrossRef]
  107. Ren, H.; Yang, W.; Jing, W.; Shahid, M.O.; Liu, Y.; Qiu, X.; Choisy, P.; Xu, T.; Ma, N.; Gao, J.; et al. Multi-omics analysis reveals key regulatory defense pathways and genes involved in salt tolerance of rose plants. Hortic. Res. 2024, 11, uhae068. [Google Scholar] [CrossRef] [PubMed]
  108. Zarour, H.M. Reversing T-cell Dysfunction and Exhaustion in Cancer. Clin. Cancer Res. 2016, 22, 1856–1864. [Google Scholar] [CrossRef] [PubMed]
  109. Peng, S.; Wang, R.; Zhang, X.; Ma, Y.; Zhong, L.; Li, K.; Nishiyama, A.; Arai, S.; Yano, S.; Wang, W. EGFR-TKI resistance promotes immune escape in lung cancer via increased PD-L1 expression. Mol. Cancer 2019, 18, 165. [Google Scholar] [CrossRef]
  110. Lee, B.S.; Park, D.I.; Lee, D.H.; Lee, J.E.; Yeo, M.K.; Park, Y.H.; Lim, D.S.; Choi, W.; Lee, D.H.; Yoo, G.; et al. Hippo effector YAP directly regulates the expression of PD-L1 transcripts in EGFR-TKI-resistant lung adenocarcinoma. Biochem. Biophys. Res. Commun. 2017, 491, 493–499. [Google Scholar] [CrossRef] [PubMed]
  111. Han, J.J.; Kim, D.W.; Koh, J.; Keam, B.; Kim, T.M.; Jeon, Y.K.; Lee, S.H.; Chung, D.H.; Heo, D.S. Change in PD-L1 Expression After Acquiring Resistance to Gefitinib in EGFR-Mutant Non-Small-Cell Lung Cancer. Clin. Lung Cancer 2016, 17, 263–270.e2. [Google Scholar] [CrossRef] [PubMed]
  112. Yang, C.Y.; Liao, W.Y.; Ho, C.C.; Chen, K.Y.; Tsai, T.H.; Hsu, C.L.; Su, K.Y.; Chang, Y.L.; Wu, C.T.; Hsu, C.C.; et al. Association between programmed death-ligand 1 expression, immune microenvironments, and clinical outcomes in epidermal growth factor receptor mutant lung adenocarcinoma patients treated with tyrosine kinase inhibitors. Eur. J. Cancer 2020, 124, 110–122. [Google Scholar] [CrossRef] [PubMed]
  113. Lasvergnas, J.; Fallet, V.; Duchemann, B.; Jouveshomme, S.; Cadranel, J.; Chouaid, C. PDL1-status predicts primary resistance of metastatic, EGFR-mutated non small cell lung cancers to EGFR tyrosine-kinase inhibitors. Respir. Med. Res. 2023, 84, 101018. [Google Scholar] [CrossRef] [PubMed]
  114. Lei, S.Y.; Xu, H.Y.; Li, H.S.; Yang, Y.N.; Xu, F.; Li, J.L.; Wang, Z.J.; Xing, P.Y.; Hao, X.Z.; Wang, Y. Influence of PD-L1 expression on the efficacy of EGFR-TKIs in EGFR-mutant non-small cell lung cancer. Thorac. Cancer 2023, 14, 2327–2337. [Google Scholar] [CrossRef]
  115. Jiang, L.; Guo, F.; Liu, X.; Li, X.; Qin, Q.; Shu, P.; Li, Y.; Wang, Y. Continuous targeted kinase inhibitors treatment induces upregulation of PD-L1 in resistant NSCLC. Sci. Rep. 2019, 9, 3705. [Google Scholar] [CrossRef]
  116. Ding, W.; Yang, P.; Zhao, X.; Wang, X.; Liu, H.; Su, Q.; Wang, X.; Li, J.; Gong, Z.; Zhang, D.; et al. Unraveling EGFR-TKI resistance in lung cancer with high PD-L1 or TMB in EGFR-sensitive mutations. Respir. Res. 2024, 25, 40. [Google Scholar] [CrossRef]
  117. Jacobsen, K.; Bertran-Alamillo, J.; Molina, M.A.; Teixido, C.; Karachaliou, N.; Pedersen, M.H.; Castellvi, J.; Garzon, M.; Codony-Servat, C.; Codony-Servat, J.; et al. Convergent Akt activation drives acquired EGFR inhibitor resistance in lung cancer. Nat. Commun. 2017, 8, 410. [Google Scholar] [CrossRef] [PubMed]
  118. Isobe, K.; Kakimoto, A.; Mikami, T.; Kaburaki, K.; Kobayashi, H.; Yoshizawa, T.; Nakano, Y.; Makino, T.; Otsuka, H.; Sano, G.; et al. PD-L1 mRNA expression in EGFR-mutant lung adenocarcinoma. Oncol. Rep. 2018, 40, 331–338. [Google Scholar] [CrossRef] [PubMed]
  119. Zheng, X.; Turkowski, K.; Mora, J.; Brune, B.; Seeger, W.; Weigert, A.; Savai, R. Redirecting tumor-associated macrophages to become tumoricidal effectors as a novel strategy for cancer therapy. Oncotarget 2017, 8, 48436–48452. [Google Scholar] [CrossRef] [PubMed]
  120. Hill, W.; Lim, E.L.; Weeden, C.E.; Lee, C.; Augustine, M.; Chen, K.; Kuan, F.C.; Marongiu, F.; Evans, E.J., Jr.; Moore, D.A.; et al. Lung adenocarcinoma promotion by air pollutants. Nature 2023, 616, 159–167. [Google Scholar] [CrossRef]
  121. Ruffell, B.; Chang-Strachan, D.; Chan, V.; Rosenbusch, A.; Ho, C.M.; Pryer, N.; Daniel, D.; Hwang, E.S.; Rugo, H.S.; Coussens, L.M. Macrophage IL-10 blocks CD8+ T cell-dependent responses to chemotherapy by suppressing IL-12 expression in intratumoral dendritic cells. Cancer Cell 2014, 26, 623–637. [Google Scholar] [CrossRef]
  122. Lin, Z.; Wang, Q.; Jiang, T.; Wang, W.; Zhao, J.J. Targeting tumor-associated macrophages with STING agonism improves the antitumor efficacy of osimertinib in a mouse model of EGFR-mutant lung cancer. Front. Immunol. 2023, 14, 1077203. [Google Scholar] [CrossRef]
  123. Peng, H.; Chen, B.; Huang, W.; Tang, Y.; Jiang, Y.; Zhang, W.; Huang, Y. Reprogramming Tumor-Associated Macrophages To Reverse EGFR(T790M) Resistance by Dual-Targeting Codelivery of Gefitinib/Vorinostat. Nano Lett. 2017, 17, 7684–7690. [Google Scholar] [CrossRef]
  124. Wang, X.; Xu, J.; Chen, J.; Jin, S.; Yao, J.; Yu, T.; Wang, W.; Guo, R. IL-22 Confers EGFR-TKI Resistance in NSCLC via the AKT and ERK Signaling Pathways. Front. Oncol. 2019, 9, 1167. [Google Scholar] [CrossRef]
  125. Hong, S.H.; Kang, N.; Kim, O.; Hong, S.A.; Park, J.; Kim, J.; Lee, M.A.; Kang, J. EGFR-Tyrosine Kinase Inhibitors Induced Activation of the Autocrine CXCL10/CXCR3 Pathway through Crosstalk between the Tumor and the Microenvironment in EGFR-Mutant Lung Cancer. Cancers 2022, 15, 124. [Google Scholar] [CrossRef] [PubMed]
  126. Li, L.; Li, Z.; Lu, C.; Li, J.; Zhang, K.; Lin, C.; Tang, X.; Liu, Z.; Zhang, Y.; Han, R.; et al. Ibrutinib reverses IL-6-induced osimertinib resistance through inhibition of Laminin alpha5/FAK signaling. Commun. Biol. 2022, 5, 155. [Google Scholar] [CrossRef]
  127. Patel, S.A.; Nilsson, M.B.; Yang, Y.; Le, X.; Tran, H.T.; Elamin, Y.Y.; Yu, X.; Zhang, F.; Poteete, A.; Ren, X.; et al. IL6 Mediates Suppression of T- and NK-cell Function in EMT-associated TKI-resistant EGFR-mutant NSCLC. Clin. Cancer Res. 2023, 29, 1292–1304. [Google Scholar] [CrossRef] [PubMed]
  128. Belluomini, L.; Cesta Incani, U.; Smimmo, A.; Avancini, A.; Sposito, M.; Insolda, J.; Mariangela Scaglione, I.; Gattazzo, F.; Caligola, S.; Adamo, A.; et al. Prognostic impact of Interleukin-8 levels in lung cancer: A meta-analysis and a bioinformatic validation. Lung Cancer 2024, 194, 107893. [Google Scholar] [CrossRef]
  129. Fernando, R.I.; Hamilton, D.H.; Dominguez, C.; David, J.M.; McCampbell, K.K.; Palena, C. IL-8 signaling is involved in resistance of lung carcinoma cells to erlotinib. Oncotarget 2016, 7, 42031–42044. [Google Scholar] [CrossRef] [PubMed]
  130. Colak, S.; Ten Dijke, P. Targeting TGF-beta Signaling in Cancer. Trends Cancer 2017, 3, 56–71. [Google Scholar] [CrossRef] [PubMed]
  131. Jiang, X.M.; Xu, Y.L.; Yuan, L.W.; Zhang, L.L.; Huang, M.Y.; Ye, Z.H.; Su, M.X.; Chen, X.P.; Zhu, H.; Ye, R.D.; et al. TGFbeta2-mediated epithelial-mesenchymal transition and NF-kappaB pathway activation contribute to osimertinib resistance. Acta Pharmacol. Sin. 2021, 42, 451–459. [Google Scholar] [CrossRef]
  132. Chen, J.; Fei, X.; Wang, J.; Cai, Z. Tumor-derived extracellular vesicles: Regulators of tumor microenvironment and the enlightenment in tumor therapy. Pharmacol. Res. 2020, 159, 105041. [Google Scholar] [CrossRef] [PubMed]
  133. Liu, J.; Ren, L.; Li, S.; Li, W.; Zheng, X.; Yang, Y.; Fu, W.; Yi, J.; Wang, J.; Du, G. The biology, function, and applications of exosomes in cancer. Acta Pharm. Sin. B 2021, 11, 2783–2797. [Google Scholar] [CrossRef]
  134. Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef] [PubMed]
  135. Zheng, B.; Song, X.; Wang, L.; Zhang, Y.; Tang, Y.; Wang, S.; Li, L.; Wu, Y.; Song, X.; Xie, L. Plasma exosomal tRNA-derived fragments as diagnostic biomarkers in non-small cell lung cancer. Front. Oncol. 2022, 12, 1037523. [Google Scholar] [CrossRef] [PubMed]
  136. Melo, S.A.; Luecke, L.B.; Kahlert, C.; Fernandez, A.F.; Gammon, S.T.; Kaye, J.; LeBleu, V.S.; Mittendorf, E.A.; Weitz, J.; Rahbari, N.; et al. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature 2015, 523, 177–182. [Google Scholar] [CrossRef] [PubMed]
  137. Jouida, A.; O’Callaghan, M.; Mc Carthy, C.; Fabre, A.; Nadarajan, P.; Keane, M.P. Exosomes from EGFR-Mutated Adenocarcinoma Induce a Hybrid EMT and MMP9-Dependant Tumor Invasion. Cancers 2022, 14, 3776. [Google Scholar] [CrossRef]
  138. Clark, D.J.; Fondrie, W.E.; Yang, A.; Mao, L. Triple SILAC quantitative proteomic analysis reveals differential abundance of cell signaling proteins between normal and lung cancer-derived exosomes. J. Proteom. 2016, 133, 161–169. [Google Scholar] [CrossRef]
  139. Park, J.O.; Choi, D.Y.; Choi, D.S.; Kim, H.J.; Kang, J.W.; Jung, J.H.; Lee, J.H.; Kim, J.; Freeman, M.R.; Lee, K.Y.; et al. Identification and characterization of proteins isolated from microvesicles derived from human lung cancer pleural effusions. Proteomics 2013, 13, 2125–2134. [Google Scholar] [CrossRef] [PubMed]
  140. Tang, S.; Yu, S.; Cheng, J.; Zhang, Y.; Huang, X. The versatile roles and clinical implications of exosomal mRNAs and microRNAs in cancer. Int. J. Biol. Markers 2020, 35, 3–19. [Google Scholar] [CrossRef]
  141. Backes, C.; Meese, E.; Keller, A. Specific miRNA Disease Biomarkers in Blood, Serum and Plasma: Challenges and Prospects. Mol. Diagn. Ther. 2016, 20, 509–518. [Google Scholar] [CrossRef] [PubMed]
  142. Peng, X.X.; Yu, R.; Wu, X.; Wu, S.Y.; Pi, C.; Chen, Z.H.; Zhang, X.C.; Gao, C.Y.; Shao, Y.W.; Liu, L.; et al. Correlation of plasma exosomal microRNAs with the efficacy of immunotherapy in EGFR/ALK wild-type advanced non-small cell lung cancer. J. Immunother. Cancer 2020, 8, e000376. [Google Scholar] [CrossRef]
  143. Liu, X.; Jiang, T.; Li, X.; Zhao, C.; Li, J.; Zhou, F.; Zhang, L.; Zhao, S.; Jia, Y.; Shi, J.; et al. Exosomes transmit T790M mutation-induced resistance in EGFR-mutant NSCLC by activating PI3K/AKT signalling pathway. J. Cell Mol. Med. 2020, 24, 1529–1540. [Google Scholar] [CrossRef] [PubMed]
  144. Yuan, S.; Chen, W.; Yang, J.; Zheng, Y.; Ye, W.; Xie, H.; Dong, L.; Xie, J. Tumor-associated macrophage-derived exosomes promote EGFR-TKI resistance in non-small cell lung cancer by regulating the AKT, ERK1/2 and STAT3 signaling pathways. Oncol. Lett. 2022, 24, 356. [Google Scholar] [CrossRef] [PubMed]
  145. Li, X.; Chen, C.; Wang, Z.; Liu, J.; Sun, W.; Shen, K.; Lv, Y.; Zhu, S.; Zhan, P.; Lv, T.; et al. Elevated exosome-derived miRNAs predict osimertinib resistance in non-small cell lung cancer. Cancer Cell Int. 2021, 21, 428. [Google Scholar] [CrossRef] [PubMed]
  146. Hisakane, K.; Seike, M.; Sugano, T.; Yoshikawa, A.; Matsuda, K.; Takano, N.; Takahashi, S.; Noro, R.; Gemma, A. Exosome-derived miR-210 involved in resistance to osimertinib and epithelial-mesenchymal transition in EGFR mutant non-small cell lung cancer cells. Thorac. Cancer 2021, 12, 1690–1698. [Google Scholar] [CrossRef] [PubMed]
  147. Takata, S.; Morikawa, K.; Tanaka, H.; Itani, H.; Ishihara, M.; Horiuchi, K.; Kato, Y.; Ikemura, S.; Nakagawa, H.; Nakahara, Y.; et al. Prospective exosome-focused translational research for afatinib (EXTRA) study of patients with nonsmall cell lung cancer harboring EGFR mutation: An observational clinical study. Ther. Adv. Med. Oncol. 2023, 15, 17588359231177021. [Google Scholar] [CrossRef]
  148. Ishii, G. New insights into cancer pathology learned from the dynamics of cancer-associated fibroblasts. Pathol. Int. 2024, 74, 493–507. [Google Scholar] [CrossRef] [PubMed]
  149. Bejarano, L.; Jordao, M.J.C.; Joyce, J.A. Therapeutic Targeting of the Tumor Microenvironment. Cancer Discov. 2021, 11, 933–959. [Google Scholar] [CrossRef]
  150. Yoshida, T.; Ishii, G.; Goto, K.; Neri, S.; Hashimoto, H.; Yoh, K.; Niho, S.; Umemura, S.; Matsumoto, S.; Ohmatsu, H.; et al. Podoplanin-positive cancer-associated fibroblasts in the tumor microenvironment induce primary resistance to EGFR-TKIs in lung adenocarcinoma with EGFR mutation. Clin. Cancer Res. 2015, 21, 642–651. [Google Scholar] [CrossRef] [PubMed]
  151. Yi, Y.; Zeng, S.; Wang, Z.; Wu, M.; Ma, Y.; Ye, X.; Zhang, B.; Liu, H. Cancer-associated fibroblasts promote epithelial-mesenchymal transition and EGFR-TKI resistance of non-small cell lung cancers via HGF/IGF-1/ANXA2 signaling. Biochim. Biophys. Acta Mol. Basis Dis. 2018, 1864, 793–803. [Google Scholar] [CrossRef] [PubMed]
  152. Li, K.; Wang, R.; Liu, G.W.; Peng, Z.Y.; Wang, J.C.; Xiao, G.D.; Tang, S.C.; Du, N.; Zhang, J.; Zhang, J.; et al. Refining the optimal CAF cluster marker for predicting TME-dependent survival expectancy and treatment benefits in NSCLC patients. Sci. Rep. 2024, 14, 16766. [Google Scholar] [CrossRef]
  153. Shintani, Y.; Kimura, T.; Funaki, S.; Ose, N.; Kanou, T.; Fukui, E. Therapeutic Targeting of Cancer-Associated Fibroblasts in the Non-Small Cell Lung Cancer Tumor Microenvironment. Cancers 2023, 15, 335. [Google Scholar] [CrossRef] [PubMed]
  154. Suzuki, E.; Yamazaki, S.; Naito, T.; Hashimoto, H.; Okubo, S.; Udagawa, H.; Goto, K.; Tsuboi, M.; Ochiai, A.; Ishii, G. Secretion of high amounts of hepatocyte growth factor is a characteristic feature of cancer-associated fibroblasts with EGFR-TKI resistance-promoting phenotype: A study of 18 cases of cancer-associated fibroblasts. Pathol. Int. 2019, 69, 472–480. [Google Scholar] [CrossRef] [PubMed]
  155. Feng, H.; Cao, B.; Peng, X.; Wei, Q. Cancer-associated fibroblasts strengthen cell proliferation and EGFR TKIs resistance through aryl hydrocarbon receptor dependent signals in non-small cell lung cancer. BMC Cancer 2022, 22, 764. [Google Scholar] [CrossRef] [PubMed]
  156. Tan, J.; Zhu, L.; Shi, J.; Zhang, J.; Kuang, J.; Guo, Q.; Zhu, X.; Chen, Y.; Zhou, C.; Gao, X. Evaluation of drug resistance for EGFR-TKIs in lung cancer via multicellular lung-on-a-chip. Eur. J. Pharm. Sci. 2024, 199, 106805. [Google Scholar] [CrossRef] [PubMed]
  157. Moghal, N.; Li, Q.; Stewart, E.L.; Navab, R.; Mikubo, M.; D’Arcangelo, E.; Martins-Filho, S.N.; Raghavan, V.; Pham, N.A.; Li, M.; et al. Single-Cell Analysis Reveals Transcriptomic Features of Drug-Tolerant Persisters and Stromal Adaptation in a Patient-Derived EGFR-Mutated Lung Adenocarcinoma Xenograft Model. J. Thorac. Oncol. 2023, 18, 499–515. [Google Scholar] [CrossRef] [PubMed]
  158. Yoshida, T.; Ishii, G.; Goto, K.; Yoh, K.; Niho, S.; Umemura, S.; Matsumoto, S.; Ohmatsu, H.; Nagai, K.; Ohe, Y.; et al. Solid predominant histology predicts EGFR tyrosine kinase inhibitor response in patients with EGFR mutation-positive lung adenocarcinoma. J. Cancer Res. Clin. Oncol. 2013, 139, 1691–1700. [Google Scholar] [CrossRef] [PubMed]
  159. Saruwatari, K.; Ikemura, S.; Sekihara, K.; Kuwata, T.; Fujii, S.; Umemura, S.; Kirita, K.; Matsumoto, S.; Yoh, K.; Niho, S.; et al. Aggressive tumor microenvironment of solid predominant lung adenocarcinoma subtype harboring with epidermal growth factor receptor mutations. Lung Cancer 2016, 91, 7–14. [Google Scholar] [CrossRef]
  160. Hu, H.; Piotrowska, Z.; Hare, P.J.; Chen, H.; Mulvey, H.E.; Mayfield, A.; Noeen, S.; Kattermann, K.; Greenberg, M.; Williams, A.; et al. Three subtypes of lung cancer fibroblasts define distinct therapeutic paradigms. Cancer Cell 2021, 39, 1531–1547.e1510. [Google Scholar] [CrossRef] [PubMed]
  161. Ballester, B.; Milara, J.; Cortijo, J. Idiopathic Pulmonary Fibrosis and Lung Cancer: Mechanisms and Molecular Targets. Int. J. Mol. Sci. 2019, 20, 593. [Google Scholar] [CrossRef] [PubMed]
  162. Kato, R.; Haratani, K.; Hayashi, H.; Sakai, K.; Sakai, H.; Kawakami, H.; Tanaka, K.; Takeda, M.; Yonesaka, K.; Nishio, K.; et al. Nintedanib promotes antitumour immunity and shows antitumour activity in combination with PD-1 blockade in mice: Potential role of cancer-associated fibroblasts. Br. J. Cancer 2021, 124, 914–924. [Google Scholar] [CrossRef] [PubMed]
  163. Fujiwara, A.; Funaki, S.; Fukui, E.; Kimura, K.; Kanou, T.; Ose, N.; Minami, M.; Shintani, Y. Effects of pirfenidone targeting the tumor microenvironment and tumor-stroma interaction as a novel treatment for non-small cell lung cancer. Sci. Rep. 2020, 10, 10900. [Google Scholar] [CrossRef]
  164. Wu, T.; Dai, Y. Tumor microenvironment and therapeutic response. Cancer Lett. 2017, 387, 61–68. [Google Scholar] [CrossRef]
  165. Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed]
  166. Saito, H.; Fukuhara, T.; Furuya, N.; Watanabe, K.; Sugawara, S.; Iwasawa, S.; Tsunezuka, Y.; Yamaguchi, O.; Okada, M.; Yoshimori, K.; et al. Erlotinib plus bevacizumab versus erlotinib alone in patients with EGFR-positive advanced non-squamous non-small-cell lung cancer (NEJ026): Interim analysis of an open-label, randomised, multicentre, phase 3 trial. Lancet Oncol. 2019, 20, 625–635. [Google Scholar] [CrossRef]
  167. Osude, C.; Lin, L.; Patel, M.; Eckburg, A.; Berei, J.; Kuckovic, A.; Dube, N.; Rastogi, A.; Gautam, S.; Smith, T.J.; et al. Mediating EGFR-TKI Resistance by VEGF/VEGFR Autocrine Pathway in Non-Small Cell Lung Cancer. Cells 2022, 11, 1694. [Google Scholar] [CrossRef] [PubMed]
  168. Sun, Y.; Niu, W.; Du, F.; Du, C.; Li, S.; Wang, J.; Li, L.; Wang, F.; Hao, Y.; Li, C.; et al. Safety, pharmacokinetics, and antitumor properties of anlotinib, an oral multi-target tyrosine kinase inhibitor, in patients with advanced refractory solid tumors. J. Hematol. Oncol. 2016, 9, 105. [Google Scholar] [CrossRef] [PubMed]
  169. Wang, G.; Sun, J.; Zhang, J.; Zhu, Q.; Lu, J.; Gao, S.; Wang, F.; Yin, Q.; Wan, Y.; Li, Q. Single-cell transcriptional profiling uncovers the association between EOMES(+)CD8(+) T cells and acquired EGFR-TKI resistance. Drug Resist. Updat. 2023, 66, 100910. [Google Scholar] [CrossRef]
  170. Hong, S.-h.; Kang, N.; Kim, S.j.; Kim, O.; Kang, J.-h.; Sung, S.W. Abstract 1192: Increased regulatory T cells induced by glycolytic metabolic change in EGFR mutant NSCLC after EGFR TKI therapy. Cancer Res. 2019, 79, 1192. [Google Scholar] [CrossRef]
  171. Liu, Y.T.; Mao, Z.W.; Ding, Y.; Wang, W.L. Macrophages as Targets in Hepatocellular Carcinoma Therapy. Mol. Cancer Ther. 2024, 23, 780–790. [Google Scholar] [CrossRef] [PubMed]
  172. Sadhukhan, P.; Seiwert, T.Y. The role of macrophages in the tumor microenvironment and tumor metabolism. Semin. Immunopathol. 2023, 45, 187–201. [Google Scholar] [CrossRef]
  173. Pan, Y.; Yu, Y.; Wang, X.; Zhang, T. Tumor-Associated Macrophages in Tumor Immunity. Front. Immunol. 2020, 11, 583084. [Google Scholar] [CrossRef]
  174. Zhang, B.; Zhang, Y.; Zhao, J.; Wang, Z.; Wu, T.; Ou, W.; Wang, J.; Yang, B.; Zhao, Y.; Rao, Z.; et al. M2-polarized macrophages contribute to the decreased sensitivity of EGFR-TKIs treatment in patients with advanced lung adenocarcinoma. Med. Oncol. 2014, 31, 127. [Google Scholar] [CrossRef] [PubMed]
  175. Rakaee, M.; Busund, L.R.; Jamaly, S.; Paulsen, E.E.; Richardsen, E.; Andersen, S.; Al-Saad, S.; Bremnes, R.M.; Donnem, T.; Kilvaer, T.K. Prognostic Value of Macrophage Phenotypes in Resectable Non-Small Cell Lung Cancer Assessed by Multiplex Immunohistochemistry. Neoplasia 2019, 21, 282–293. [Google Scholar] [CrossRef] [PubMed]
  176. Wang, L.; Cao, L.; Wang, H.; Liu, B.; Zhang, Q.; Meng, Z.; Wu, X.; Zhou, Q.; Xu, K. Cancer-associated fibroblasts enhance metastatic potential of lung cancer cells through IL-6/STAT3 signaling pathway. Oncotarget 2017, 8, 76116–76128. [Google Scholar] [CrossRef] [PubMed]
  177. Yuan, A.; Chen, J.J.; Yao, P.L.; Yang, P.C. The role of interleukin-8 in cancer cells and microenvironment interaction. Front. Biosci. 2005, 10, 853–865. [Google Scholar] [CrossRef] [PubMed]
  178. Favaro, F.; Luciano-Mateo, F.; Moreno-Caceres, J.; Hernandez-Madrigal, M.; Both, D.; Montironi, C.; Puschel, F.; Nadal, E.; Eldering, E.; Munoz-Pinedo, C. TRAIL receptors promote constitutive and inducible IL-8 secretion in non-small cell lung carcinoma. Cell Death Dis. 2022, 13, 1046. [Google Scholar] [CrossRef]
  179. Liu, Y.N.; Chang, T.H.; Tsai, M.F.; Wu, S.G.; Tsai, T.H.; Chen, H.Y.; Yu, S.L.; Yang, J.C.; Shih, J.Y. IL-8 confers resistance to EGFR inhibitors by inducing stem cell properties in lung cancer. Oncotarget 2015, 6, 10415–10431. [Google Scholar] [CrossRef] [PubMed]
  180. Chang, C.F.; Westbrook, R.; Ma, J.; Cao, D. Transforming growth factor-beta signaling in breast cancer. Front. Biosci. 2007, 12, 4393–4401. [Google Scholar] [CrossRef]
  181. Kelly, R.J.; Morris, J.C. Transforming growth factor-beta: A target for cancer therapy. J. Immunotoxicol. 2010, 7, 15–26. [Google Scholar] [CrossRef] [PubMed]
  182. Liu, Z.; Han, C.; Dong, C.; Shen, A.; Hsu, E.; Ren, Z.; Lu, C.; Liu, L.; Zhang, A.; Timmerman, C.; et al. Hypofractionated EGFR tyrosine kinase inhibitor limits tumor relapse through triggering innate and adaptive immunity. Sci. Immunol. 2019, 4, eaav6473. [Google Scholar] [CrossRef]
  183. Zhou, S.; Gu, L.; He, J.; Zhang, H.; Zhou, M. MDM2 regulates vascular endothelial growth factor mRNA stabilization in hypoxia. Mol. Cell Biol. 2011, 31, 4928–4937. [Google Scholar] [CrossRef] [PubMed]
  184. Li, Z.; Rana, T.M. Decoding the noncoding: Prospective of lncRNA-mediated innate immune regulation. RNA Biol. 2014, 11, 979–985. [Google Scholar] [CrossRef] [PubMed]
  185. Wang, X.; Freire Valls, A.; Schermann, G.; Shen, Y.; Moya, I.M.; Castro, L.; Urban, S.; Solecki, G.M.; Winkler, F.; Riedemann, L.; et al. YAP/TAZ Orchestrate VEGF Signaling during Developmental Angiogenesis. Dev. Cell 2017, 42, 462–478.e467. [Google Scholar] [CrossRef]
  186. Pulkkinen, H.H.; Kiema, M.; Lappalainen, J.P.; Toropainen, A.; Beter, M.; Tirronen, A.; Holappa, L.; Niskanen, H.; Kaikkonen, M.U.; Ylä-Herttuala, S.; et al. BMP6/TAZ-Hippo signaling modulates angiogenesis and endothelial cell response to VEGF. Angiogenesis 2021, 24, 129–144. [Google Scholar] [CrossRef]
  187. Le, X.; Nilsson, M.; Goldman, J.; Reck, M.; Nakagawa, K.; Kato, T.; Ares, L.P.; Frimodt-Moller, B.; Wolff, K.; Visseren-Grul, C.; et al. Dual EGFR-VEGF Pathway Inhibition: A Promising Strategy for Patients with EGFR-Mutant NSCLC. J. Thorac. Oncol. 2021, 16, 205–215. [Google Scholar] [CrossRef]
  188. Hung, M.S.; Chen, I.C.; Lin, P.Y.; Lung, J.H.; Li, Y.C.; Lin, Y.C.; Yang, C.T.; Tsai, Y.H. Epidermal growth factor receptor mutation enhances expression of vascular endothelial growth factor in lung cancer. Oncol. Lett. 2016, 12, 4598–4604. [Google Scholar] [CrossRef] [PubMed]
  189. Motz, G.T.; Coukos, G. The parallel lives of angiogenesis and immunosuppression: Cancer and other tales. Nat. Rev. Immunol. 2011, 11, 702–711. [Google Scholar] [CrossRef] [PubMed]
  190. Zhang, Y.; Brekken, R.A. Direct and indirect regulation of the tumor immune microenvironment by VEGF. J. Leukoc. Biol. 2022, 111, 1269–1286. [Google Scholar] [CrossRef]
  191. Courau, T.; Nehar-Belaid, D.; Florez, L.; Levacher, B.; Vazquez, T.; Brimaud, F.; Bellier, B.; Klatzmann, D. TGF-β and VEGF cooperatively control the immunotolerant tumor environment and the efficacy of cancer immunotherapies. JCI Insight 2016, 1, e85974. [Google Scholar] [CrossRef]
  192. Geindreau, M.; Ghiringhelli, F.; Bruchard, M. Vascular Endothelial Growth Factor, a Key Modulator of the Anti-Tumor Immune Response. Int. J. Mol. Sci. 2021, 22, 4871. [Google Scholar] [CrossRef] [PubMed]
  193. Lee, W.S.; Yang, H.; Chon, H.J.; Kim, C. Combination of anti-angiogenic therapy and immune checkpoint blockade normalizes vascular-immune crosstalk to potentiate cancer immunity. Exp. Mol. Med. 2020, 52, 1475–1485. [Google Scholar] [CrossRef] [PubMed]
  194. Zhao, Y.; Guo, S.; Deng, J.; Shen, J.; Du, F.; Wu, X.; Chen, Y.; Li, M.; Chen, M.; Li, X.; et al. VEGF/VEGFR-Targeted Therapy and Immunotherapy in Non-small Cell Lung Cancer: Targeting the Tumor Microenvironment. Int. J. Biol. Sci. 2022, 18, 3845–3858. [Google Scholar] [CrossRef]
  195. Dong, Z.Y.; Zhang, J.T.; Liu, S.Y.; Su, J.; Zhang, C.; Xie, Z.; Zhou, Q.; Tu, H.Y.; Xu, C.R.; Yan, L.X.; et al. EGFR mutation correlates with uninflamed phenotype and weak immunogenicity, causing impaired response to PD-1 blockade in non-small cell lung cancer. Oncoimmunology 2017, 6, e1356145. [Google Scholar] [CrossRef] [PubMed]
  196. Offin, M.; Rizvi, H.; Tenet, M.; Ni, A.; Sanchez-Vega, F.; Li, B.T.; Drilon, A.; Kris, M.G.; Rudin, C.M.; Schultz, N.; et al. Tumor Mutation Burden and Efficacy of EGFR-Tyrosine Kinase Inhibitors in Patients with EGFR-Mutant Lung Cancers. Clin. Cancer Res. 2019, 25, 1063–1069. [Google Scholar] [CrossRef] [PubMed]
  197. Yoneshima, Y.; Ijichi, K.; Anai, S.; Ota, K.; Otsubo, K.; Iwama, E.; Tanaka, K.; Oda, Y.; Nakanishi, Y.; Okamoto, I. PD-L1 expression in lung adenocarcinoma harboring EGFR mutations or ALK rearrangements. Lung Cancer 2018, 118, 36–40. [Google Scholar] [CrossRef]
  198. Yoon, B.W.; Chang, B.; Lee, S.H. High PD-L1 Expression is Associated with Unfavorable Clinical Outcome in EGFR-Mutated Lung Adenocarcinomas Treated with Targeted Therapy. Onco Targets Ther. 2020, 13, 8273–8285. [Google Scholar] [CrossRef] [PubMed]
  199. Soo, R.A.; Kim, H.R.; Asuncion, B.R.; Fazreen, Z.; Omar, M.F.M.; Herrera, M.C.; Yun Lim, J.S.; Sia, G.; Soong, R.; Cho, B.C. Significance of immune checkpoint proteins in EGFR-mutant non-small cell lung cancer. Lung Cancer 2017, 105, 17–22. [Google Scholar] [CrossRef]
  200. Hsu, K.H.; Huang, Y.H.; Tseng, J.S.; Chen, K.C.; Ku, W.H.; Su, K.Y.; Chen, J.J.W.; Chen, H.W.; Yu, S.L.; Yang, T.Y.; et al. High PD-L1 expression correlates with primary resistance to EGFR-TKIs in treatment naive advanced EGFR-mutant lung adenocarcinoma patients. Lung Cancer 2019, 127, 37–43. [Google Scholar] [CrossRef] [PubMed]
  201. Wang, Y.; Zhang, T.; Guo, L.; Ren, T.; Yang, Y. Stromal extracellular matrix is a microenvironmental cue promoting resistance to EGFR tyrosine kinase inhibitors in lung cancer cells. Int. J. Biochem. Cell Biol. 2019, 106, 96–106. [Google Scholar] [CrossRef]
  202. Mezu-Ndubuisi, O.J.; Maheshwari, A. The role of integrins in inflammation and angiogenesis. Pediatr. Res. 2021, 89, 1619–1626. [Google Scholar] [CrossRef] [PubMed]
  203. Liu, F.; Wu, Q.; Dong, Z.; Liu, K. Integrins in cancer: Emerging mechanisms and therapeutic opportunities. Pharmacol. Ther. 2023, 247, 108458. [Google Scholar] [CrossRef] [PubMed]
  204. Bou Antoun, N.; Chioni, A.-M. Dysregulated Signalling Pathways Driving Anticancer Drug Resistance. Int. J. Mol. Sci. 2023, 24, 12222. [Google Scholar] [CrossRef]
  205. Wang, C.; Wang, T.; Lv, D.; Li, L.; Yue, J.; Chen, H.Z.; Xu, L. Acquired Resistance to EGFR TKIs Mediated by TGFbeta1/Integrin beta3 Signaling in EGFR-Mutant Lung Cancer. Mol. Cancer Ther. 2019, 18, 2357–2367. [Google Scholar] [CrossRef] [PubMed]
  206. Yamazaki, S.; Higuchi, Y.; Ishibashi, M.; Hashimoto, H.; Yasunaga, M.; Matsumura, Y.; Tsuchihara, K.; Tsuboi, M.; Goto, K.; Ochiai, A.; et al. Collagen type I induces EGFR-TKI resistance in EGFR-mutated cancer cells by mTOR activation through Akt-independent pathway. Cancer Sci. 2018, 109, 2063–2073. [Google Scholar] [CrossRef] [PubMed]
  207. Vigano, S.; Alatzoglou, D.; Irving, M.; Ménétrier-Caux, C.; Caux, C.; Romero, P.; Coukos, G. Targeting adenosine in cancer immunotherapy to enhance T-cell function. Front. Immunol. 2019, 10, 925. [Google Scholar] [CrossRef] [PubMed]
  208. Passarelli, A.; Tucci, M.; Mannavola, F.; Felici, C.; Silvestris, F. The metabolic milieu in melanoma: Role of immune suppression by CD73/adenosine. Tumor Biol. 2019, 41, 1010428319837138. [Google Scholar] [CrossRef]
  209. Fong, L.; Forde, P.M.; Powderly, J.D.; Goldman, J.W.; Nemunaitis, J.J.; Luke, J.J.; Hellmann, M.D.; Kummar, S.; Doebele, R.C.; Mahadevan, D. Safety and clinical activity of adenosine A2a receptor (A2aR) antagonist, CPI-444, in anti-PD1/PDL1 treatment-refractory renal cell (RCC) and non-small cell lung cancer (NSCLC) patients. J. Clin. Oncol. 2017, 35, 3004. [Google Scholar] [CrossRef]
  210. Passarelli, A.; Aieta, M.; Sgambato, A.; Gridelli, C. Targeting Immunometabolism Mediated by CD73 Pathway in EGFR-Mutated Non-small Cell Lung Cancer: A New Hope for Overcoming Immune Resistance. Front. Immunol. 2020, 11, 1479. [Google Scholar] [CrossRef]
  211. Le, X.; Negrao, M.V.; Reuben, A.; Federico, L.; Diao, L.; McGrail, D.; Nilsson, M.; Robichaux, J.; Munoz, I.G.; Patel, S.; et al. Characterization of the Immune Landscape of EGFR-Mutant NSCLC Identifies CD73/Adenosine Pathway as a Potential Therapeutic Target. J. Thorac. Oncol. 2021, 16, 583–600. [Google Scholar] [CrossRef]
  212. Tu, E.; McGlinchey, K.; Wang, J.; Martin, P.; Ching, S.L.K.; Floc’h, N.; Kurasawa, J.; Starrett, J.H.; Lazdun, Y.; Wetzel, L.; et al. Anti–PD-L1 and anti-CD73 combination therapy promotes T cell response to EGFR-mutated NSCLC. JCI Insight 2022, 7, e142843. [Google Scholar] [CrossRef] [PubMed]
  213. Qin, W.; Zou, J.; Huang, Y.; Liu, C.; Kang, Y.; Han, H.; Tang, Y.; Li, L.; Liu, B.; Zhao, W.; et al. Pirfenidone facilitates immune infiltration and enhances the antitumor efficacy of PD-L1 blockade in mice. Oncoimmunology 2020, 9, 1824631. [Google Scholar] [CrossRef]
  214. Corrales, L.; Nogueira, A.; Passiglia, F.; Listi, A.; Caglevic, C.; Giallombardo, M.; Raez, L.; Santos, E.; Rolfo, C. Second-Line Treatment of Non-Small Cell Lung Cancer: Clinical, Pathological, and Molecular Aspects of Nintedanib. Front. Med. 2017, 4, 13. [Google Scholar] [CrossRef]
  215. Yan, S.; Xue, S.; Wang, T.; Gao, R.; Zeng, H.; Wang, Q.; Jia, X. Efficacy and safety of nintedanib in patients with non-small cell lung cancer, and novel insights in radiation-induced lung toxicity. Front. Oncol. 2023, 13, 1086214. [Google Scholar] [CrossRef] [PubMed]
  216. Epstein Shochet, G.; Israeli-Shani, L.; Koslow, M.; Shitrit, D. Nintedanib (BIBF 1120) blocks the tumor promoting signals of lung fibroblast soluble microenvironment. Lung Cancer 2016, 96, 7–14. [Google Scholar] [CrossRef]
  217. Rannikko, J.H.; Hollmen, M. Clinical landscape of macrophage-reprogramming cancer immunotherapies. Br. J. Cancer 2024, 131, 627–640. [Google Scholar] [CrossRef] [PubMed]
  218. Yin, W.; Yu, X.; Kang, X.; Zhao, Y.; Zhao, P.; Jin, H.; Fu, X.; Wan, Y.; Peng, C.; Huang, Y. Remodeling Tumor-Associated Macrophages and Neovascularization Overcomes EGFRT790M-Associated Drug Resistance by PD-L1 Nanobody-Mediated Codelivery. Small 2018, 14, 1802372. [Google Scholar] [CrossRef]
  219. Chen, M.T.; Li, B.Z.; Zhang, E.P.; Zheng, Q. Potential roles of tumor microenvironment in gefitinib-resistant non-small cell lung cancer: A narrative review. Medicine 2023, 102, e35086. [Google Scholar] [CrossRef]
  220. Yang, J.C.-H.; Lee, D.H.; Lee, J.-S.; Fan, Y.; Marinis, F.d.; Iwama, E.; Inoue, T.; Rodríguez-Cid, J.; Zhang, L.; Yang, C.-T.; et al. Phase III KEYNOTE-789 Study of Pemetrexed and Platinum with or Without Pembrolizumab for Tyrosine Kinase Inhibitor-Resistant, EGFR-Mutant, Metastatic Nonsquamous Non-Small Cell Lung Cancer. J. Clin. Oncol. 2023, 42, 4029–4039. [Google Scholar] [CrossRef]
  221. National Library of Medicine. Tocilizumab and Atezolizumab in Adults with Locally Advanced or Metastatic Non-Small Cell Lung Cancer Refractory to 1st Line Immune Checkpoint Inhibitor-Based Therapy. Available online: https://clinicaltrials.gov/study/NCT04691817?tab=history (accessed on 7 October 2024).
  222. Jiang, T.; Wang, P.; Zhang, J.; Zhao, Y.; Zhou, J.; Fan, Y.; Shu, Y.; Liu, X.; Zhang, H.; He, J.; et al. Toripalimab plus chemotherapy as second-line treatment in previously EGFR-TKI treated patients with EGFR-mutant-advanced NSCLC: A multicenter phase-II trial. Signal Transduct. Target. Ther. 2021, 6, 355. [Google Scholar] [CrossRef]
  223. Garassino, M.C.; Cho, B.-C.; Kim, J.-H.; Mazières, J.; Vansteenkiste, J.; Lena, H.; Jaime, J.C.; Gray, J.E.; Powderly, J.; Chouaid, C.; et al. Final overall survival and safety update for durvalumab in third- or later-line advanced NSCLC: The phase II ATLANTIC study. Lung Cancer 2020, 147, 137–142. [Google Scholar] [CrossRef] [PubMed]
  224. Garassino, M.C.; Cho, B.-C.; Kim, J.-H.; Mazières, J.; Vansteenkiste, J.; Lena, H.; Corral Jaime, J.; Gray, J.E.; Powderly, J.; Chouaid, C.; et al. Durvalumab as third-line or later treatment for advanced non-small-cell lung cancer (ATLANTIC): An open-label, single-arm, phase 2 study. Lancet Oncol. 2018, 19, 521–536. [Google Scholar] [CrossRef]
  225. Qin, B.-D.; Jiao, X.-D.; Yuan, L.-Y.; Wu, Y.; Ling, Y.; Zang, Y.-S. Immunotherapy-based regimens for patients with EGFR-mutated non-small cell lung cancer who progressed on EGFR-TKI therapy. J. Immunother. Cancer 2024, 12, e008818. [Google Scholar] [CrossRef]
  226. Lu, S.; Wu, L.; Jian, H.; Chen, Y.; Wang, Q.; Fang, J.; Wang, Z.; Hu, Y.; Sun, M.; Han, L.; et al. Sintilimab plus bevacizumab biosimilar IBI305 and chemotherapy for patients with EGFR-mutated non-squamous non-small-cell lung cancer who progressed on EGFR tyrosine-kinase inhibitor therapy (ORIENT-31): First interim results from a randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol. 2022, 23, 1167–1179. [Google Scholar] [CrossRef] [PubMed]
  227. Yang, J.C.-H.; Lee, D.H.; Lee, J.-S.; Fan, Y.; Marinis, F.d.; Okamoto, I.; Inoue, T.; Cid, J.R.R.; Zhang, L.; Yang, C.-T.; et al. Pemetrexed and platinum with or without pembrolizumab for tyrosine kinase inhibitor (TKI)-resistant, EGFR-mutant, metastatic nonsquamous NSCLC: Phase 3 KEYNOTE-789 study. J. Clin. Oncol. 2023, 41, LBA9000. [Google Scholar] [CrossRef]
  228. Min, J.W.; Lim, J.U. Immune checkpoint inhibitors in patients with chronic kidney disease: Assessing their ability to cause acute kidney injury and informing their proper use. Semin. Oncol. 2022, 49, 141–147. [Google Scholar] [CrossRef] [PubMed]
  229. Socinski, M.A.; Jotte, R.M.; Cappuzzo, F.; Nishio, M.; Mok, T.S.K.; Reck, M.; Finley, G.G.; Kaul, M.D.; Yu, W.; Paranthaman, N.; et al. Association of Immune-Related Adverse Events with Efficacy of Atezolizumab in Patients with Non–Small Cell Lung Cancer: Pooled Analyses of the Phase 3 IMpower130, IMpower132, and IMpower150 Randomized Clinical Trials. JAMA Oncol. 2023, 9, 527–535. [Google Scholar] [CrossRef]
  230. Yan, D. Hope and Challenges: Immunotherapy in EGFR-Mutant NSCLC Patients. Biomedicines 2023, 11, 2916. [Google Scholar] [CrossRef]
  231. Liu, S.; Wu, F.; Li, X.; Zhao, C.; Jia, Y.; Jia, K.; Han, R.; Qiao, M.; Li, W.; Yu, J.; et al. Patients with Short PFS to EGFR-TKIs Predicted Better Response to Subsequent Anti-PD-1/PD-L1 Based Immunotherapy in EGFR Common Mutation NSCLC. Front. Oncol. 2021, 11, 639947. [Google Scholar] [CrossRef]
  232. Lisberg, A.; Cummings, A.; Goldman, J.W.; Bornazyan, K.; Reese, N.; Wang, T.; Coluzzi, P.; Ledezma, B.; Mendenhall, M.; Hunt, J.; et al. A Phase II Study of Pembrolizumab in EGFR-Mutant, PD-L1+, Tyrosine Kinase Inhibitor Naïve Patients with Advanced NSCLC. J. Thorac. Oncol. 2018, 13, 1138–1145. [Google Scholar] [CrossRef]
  233. Nakagawa, K.; Garon, E.B.; Seto, T.; Nishio, M.; Ponce Aix, S.; Paz-Ares, L.; Chiu, C.-H.; 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 randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2019, 20, 1655–1669. [Google Scholar] [CrossRef]
  234. Yu, L.; Hu, Y.; Xu, J.; Qiao, R.; Zhong, H.; Han, B.; Xia, J.; Zhong, R. Multi-target angiogenesis inhibitor combined with PD-1 inhibitors may benefit advanced non-small cell lung cancer patients in late line after failure of EGFR-TKI therapy. Int. J. Cancer 2023, 153, 635–643. [Google Scholar] [CrossRef] [PubMed]
  235. Reck, M.; Mok, T.S.K.; Nishio, M.; Jotte, R.M.; Cappuzzo, F.; Orlandi, F.; Stroyakovskiy, D.; Nogami, N.; Rodriguez-Abreu, D.; Moro-Sibilot, D.; et al. Atezolizumab plus bevacizumab and chemotherapy in non-small-cell lung cancer (IMpower150): Key subgroup analyses of patients with EGFR mutations or baseline liver metastases in a randomised, open-label phase 3 trial. Lancet Respir. Med. 2019, 7, 387–401. [Google Scholar] [CrossRef] [PubMed]
  236. Nogami, N.; Barlesi, F.; Socinski, M.A.; Reck, M.; Thomas, C.A.; Cappuzzo, F.; Mok, T.S.K.; Finley, G.; Aerts, J.G.; Orlandi, F.; et al. IMpower150 Final Exploratory Analyses for Atezolizumab Plus Bevacizumab and Chemotherapy in Key NSCLC Patient Subgroups with EGFR Mutations or Metastases in the Liver or Brain. J. Thorac. Oncol. 2022, 17, 309–323. [Google Scholar] [CrossRef] [PubMed]
  237. Investigators, H.A.-A.S.; Fang, W.; Zhao, Y.; Luo, Y.; Yang, R.; Huang, Y.; He, Z.; Zhao, H.; Li, M.; Li, K.; et al. Ivonescimab Plus Chemotherapy in Non-Small Cell Lung Cancer with EGFR Variant: A Randomized Clinical Trial. JAMA 2024, 332, 561–570. [Google Scholar] [CrossRef] [PubMed]
  238. Vyse, S.; Huang, P.H. Amivantamab for the treatment of EGFR exon 20 insertion mutant non-small cell lung cancer. Expert. Rev. Anticancer. Ther. 2022, 22, 3–16. [Google Scholar] [CrossRef] [PubMed]
  239. Zavaleta-Monestel, E.; Garcia-Montero, J.; Arguedas-Chacon, S.; Quesada-Villasenor, R.; Barrantes-Lopez, M.; Arroyo-Solis, R.; Zuniga-Orlich, C.E. Amivantamab: A Novel Advance in the Treatment of Non-small Cell Lung Cancer. Cureus 2024, 16, e60851. [Google Scholar] [CrossRef] [PubMed]
  240. Rivera-Soto, R.; Henley, B.; Pulgar, M.A.; Lehman, S.L.; Gupta, H.; Perez-Vale, K.Z.; Weindorfer, M.; Vijayaraghavan, S.; Yao, T.S.; Laquerre, S.; et al. Amivantamab efficacy in wild-type EGFR NSCLC tumors correlates with levels of ligand expression. NPJ Precis. Oncol. 2024, 8, 192. [Google Scholar] [CrossRef] [PubMed]
  241. Billowria, K.; Das Gupta, G.; Chawla, P.A. Amivantamab: A New Hope in Targeting Non-small Cell Lung Cancer. Anticancer. Agents Med. Chem. 2023, 23, 124–141. [Google Scholar] [CrossRef] [PubMed]
  242. Vijayaraghavan, S.; Lipfert, L.; Chevalier, K.; Bushey, B.S.; Henley, B.; Lenhart, R.; Sendecki, J.; Beqiri, M.; Millar, H.J.; Packman, K.; et al. Amivantamab (JNJ-61186372), an Fc Enhanced EGFR/cMet Bispecific Antibody, Induces Receptor Downmodulation and Antitumor Activity by Monocyte/Macrophage Trogocytosis. Mol. Cancer Ther. 2020, 19, 2044–2056. [Google Scholar] [CrossRef]
  243. 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]
  244. Kawamura, T.; Hata, A.; Takeshita, J.; Fujita, S.; Hayashi, M.; Tomii, K.; Katakami, N. High-dose erlotinib for refractory leptomeningeal metastases after failure of standard-dose EGFR-TKIs. Cancer Chemother. Pharmacol. 2015, 75, 1261–1266. [Google Scholar] [CrossRef]
  245. Gerritse, S.L.; Janssen, J.B.E.; Labots, M.; de Vries, R.; Rudek, M.; Carducci, M.; van Erp, N.P.; Verheul, H.M.W. High-dose administration of tyrosine kinase inhibitors to improve clinical benefit: A systematic review. Cancer Treat. Rev. 2021, 97, 102171. [Google Scholar] [CrossRef]
  246. Yu, D.; Kane, M.J.; Koay, E.J.; Wistuba, I.I.; Hobbs, B.P. Machine learning identifies prognostic subtypes of the tumor microenvironment of NSCLC. Sci. Rep. 2024, 14, 15004. [Google Scholar] [CrossRef] [PubMed]
  247. Chen, H.; Yang, W.; Ji, Z. Machine learning-based identification of tumor-infiltrating immune cell-associated model with appealing implications in improving prognosis and immunotherapy response in bladder cancer patients. Front. Immunol. 2023, 14, 1171420. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of key components in the tumor microenvironment (TME) of epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer (NSCLC). TME elements contributing to EGFR tyrosine kinase inhibitor (TKI) resistance. CD8+ T-cells are shown with reduced infiltration, with a specific subset (EOMES+) marked to indicate functional relevance. CD4+ T-cells (Tregs) are depicted with an increased presence near tumor cells, actively suppressing immune responses. Tumor-associated macrophages (TAMs) are illustrated with a transition from M1 (anti-tumor) to M2 (pro-tumor) phenotype, characterized by markers such as IDO1+. Cancer-associated fibroblasts (CAFs) in the stroma secrete factors such as hepatocyte growth factor (HGF) and IL-6 to promote epithelial-mesenchymal transition (EMT) and resistance in adjacent cancer cells. Cytokines, including IL-6, IL-8, and TGF-β, are shown as arrows, indicating their role in tumor growth and immune suppression. Exosomes are depicted as transferring resistance-related miRNAs between tumor and stromal cells. Abnormalities in blood vessels, i.e., leaky blood vessels, driven by vascular endothelial growth factor (VEGF) promote immunosuppressive effects. PD-L1 expression on tumor cells is elevated, inhibiting cluster of differentiation 8 (CD8+) T-cell activity and contributing to immune evasion. This schematic integrates the complex interplay of immune cells, stromal elements, and molecular signals that underpin resistance mechanisms in EGFR-mutant NSCLC. Integrins, a core component of the extracellular matrix, engage with collagen-rich ECM to promote cell adhesion and trigger alternative signaling cascades such as FAK/Src and PI3K/AKT, allowing tumor cells to proliferate. Adenosine is a key modulator of immune responses. Its accumulation is facilitated by ectonucleotidase CD73, which promotes adenosine production and subsequently activates immunosuppressive pathways in tumor-infiltrating immune cells.
Figure 1. Schematic representation of key components in the tumor microenvironment (TME) of epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer (NSCLC). TME elements contributing to EGFR tyrosine kinase inhibitor (TKI) resistance. CD8+ T-cells are shown with reduced infiltration, with a specific subset (EOMES+) marked to indicate functional relevance. CD4+ T-cells (Tregs) are depicted with an increased presence near tumor cells, actively suppressing immune responses. Tumor-associated macrophages (TAMs) are illustrated with a transition from M1 (anti-tumor) to M2 (pro-tumor) phenotype, characterized by markers such as IDO1+. Cancer-associated fibroblasts (CAFs) in the stroma secrete factors such as hepatocyte growth factor (HGF) and IL-6 to promote epithelial-mesenchymal transition (EMT) and resistance in adjacent cancer cells. Cytokines, including IL-6, IL-8, and TGF-β, are shown as arrows, indicating their role in tumor growth and immune suppression. Exosomes are depicted as transferring resistance-related miRNAs between tumor and stromal cells. Abnormalities in blood vessels, i.e., leaky blood vessels, driven by vascular endothelial growth factor (VEGF) promote immunosuppressive effects. PD-L1 expression on tumor cells is elevated, inhibiting cluster of differentiation 8 (CD8+) T-cell activity and contributing to immune evasion. This schematic integrates the complex interplay of immune cells, stromal elements, and molecular signals that underpin resistance mechanisms in EGFR-mutant NSCLC. Integrins, a core component of the extracellular matrix, engage with collagen-rich ECM to promote cell adhesion and trigger alternative signaling cascades such as FAK/Src and PI3K/AKT, allowing tumor cells to proliferate. Adenosine is a key modulator of immune responses. Its accumulation is facilitated by ectonucleotidase CD73, which promotes adenosine production and subsequently activates immunosuppressive pathways in tumor-infiltrating immune cells.
Biomedicines 13 00470 g001
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

Lim, J.U.; Jung, J.; Kim, Y.W.; Kim, C.Y.; Lee, S.H.; Park, D.W.; Choi, S.I.; Ji, W.; Yeo, C.D.; Lee, S.H. Targeting the Tumor Microenvironment in EGFR-Mutant Lung Cancer: Opportunities and Challenges. Biomedicines 2025, 13, 470. https://doi.org/10.3390/biomedicines13020470

AMA Style

Lim JU, Jung J, Kim YW, Kim CY, Lee SH, Park DW, Choi SI, Ji W, Yeo CD, Lee SH. Targeting the Tumor Microenvironment in EGFR-Mutant Lung Cancer: Opportunities and Challenges. Biomedicines. 2025; 13(2):470. https://doi.org/10.3390/biomedicines13020470

Chicago/Turabian Style

Lim, Jeong Uk, Junyang Jung, Yeon Wook Kim, Chi Young Kim, Sang Hoon Lee, Dong Won Park, Sue In Choi, Wonjun Ji, Chang Dong Yeo, and Seung Hyeun Lee. 2025. "Targeting the Tumor Microenvironment in EGFR-Mutant Lung Cancer: Opportunities and Challenges" Biomedicines 13, no. 2: 470. https://doi.org/10.3390/biomedicines13020470

APA Style

Lim, J. U., Jung, J., Kim, Y. W., Kim, C. Y., Lee, S. H., Park, D. W., Choi, S. I., Ji, W., Yeo, C. D., & Lee, S. H. (2025). Targeting the Tumor Microenvironment in EGFR-Mutant Lung Cancer: Opportunities and Challenges. Biomedicines, 13(2), 470. https://doi.org/10.3390/biomedicines13020470

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