Clinical advances in EGFR-TKI combination therapy for EGFR-mutated NSCLC: a narrative review
Review Article

Clinical advances in EGFR-TKI combination therapy for EGFR-mutated NSCLC: a narrative review

Qianru Zhang, Ruo Wang, Lu Xu

Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China

Contributions: (I) Conception and design: L Xu; (I) Administrative support: All authors; (II) Provision of study materials or patients: None; (III) Collection and assembly of data: Q Zhang; (IV) Data analysis and interpretation: Q Zhang, R Wang; (V) Manuscript writing: All authors; (VI) Final approval of manuscript: All authors.

Correspondence to: Lu Xu, PhD. Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 227 Chongqing South Road, Ruijin Second Road Street, Huangpu District, Shanghai 200025, China. Email: luxupharm@163.com.

Background and Objective: Mutations located in epidermal growth factor receptor (EGFR) tyrosine kinase domains have been described as the ‘Achilles heel’ of non-small cell lung cancer (NSCLC) and can be targeted by epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs). However, the clinical benefits of EGFR-TKIs are limited, and drug resistance inevitably occurs in NSCLC patients after long-term exposure to certain drugs. EGFR-TKI combination therapies, including combined targeted therapy, radiotherapy, chemotherapy, and immunotherapy, have shown promise in addressing this issue. This literature review analyzed the rationale and controversies of clinical research related to various EGFR-TKI combination therapies.

Methods: The PubMed database was searched to retrieve articles published from January 1, 2001 to April 15, 2023 using the following Medical Subject Headings (MeSH) terms: “EGFR-mutated non-small cell lung cancer” and “clinical trial”. Google Scholar was also reviewed to retrieve additional articles. The search was limited to articles published in English.

Key Content and Findings: In this review, we summarized EGFR-TKI combination therapies, including combined targeted therapy, radiotherapy, chemotherapy, and immunotherapy, most of which have shown efficacy and safety in patients with EGFR-mutated NSCLC. A number of clinical studies with large sample sizes have analyzed the activity and toxicity of combined therapies and explored potential and well-tolerated treatment options.

Conclusions: EGFR mutations have been detected in many NSCLC patients and can be targeted by EGFR-TKIs. However, drug resistance after long-term exposure remains a significant challenge for this type of treatment. Most clinical trials have shown that the combination of EGFR-TKIs and targeted therapy, chemotherapy, radiotherapy or immunotherapy is efficacious and safe in the treatment of EGFR-mutated NSCLC. It should be noted that in some instances, serious adverse events have led to the termination of trials. However, EGFR-TKI combination therapy is indeed an effective approach for the treatment of patients with EGFR-mutated NSCLC and deserves further development.

Keywords: Combination therapy; epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs); non-small cell lung cancer (NSCLC)


Submitted Jun 02, 2023. Accepted for publication Sep 13, 2023. Published online Nov 24, 2023.

doi: 10.21037/tcr-23-956


Introduction

Lung cancer, which is one of the most common types of cancer, remains the leading cause of cancer-related deaths in both men and women (1). In 2022, there were approximately 2.21 million new diagnoses of lung cancer (accounting for 11.4% of all cancer cases) and 1.79 million deaths caused by lung cancer worldwide (2). Non-small cell lung cancer (NSCLC) is the most common form of lung cancer, accounting for about 80–85% of all lung cancers (3). Depending on the stage of the cancer, radical surgery, platinum-based chemotherapy, radiotherapy, and immunotherapy can be applied in the treatment of NSCLC (4).

In-depth research on the oncogenesis and progression of NSCLC has shown that most NSCLC patients have mutations of the epidermal growth factor receptor (EGFR) (5). EGFR is a kind of transmembrane tyrosine kinase protein and is becoming a landmark target in the treatment of NSCLC. EGFR is activated by receptor overexpression, a common phenomenon in various cancer tissues. EGFR overexpression, it has been reported to be associated with higher aggressiveness and poorer clinical outcomes in breast, lung, ovarian, cervical, bladder, esophageal, brain, head, and neck cancers (6-10). Additionally, EGFR activation and phosphorylation by the binding of the ligands, such as epidermal growth factor (EGF) (11), further activate several downstream signaling pathways, such as the rat sarcoma protein (Ras)/rapidly accelerated fibrosarcoma protein (Raf)/mitogen-activated protein kinases (MAPK), phosphatidylinositol-3 kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR), Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathways (12), which play an important role in regulating multiple cellular processes, including proliferation, survival, and apoptosis.

Epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs), which target the EGFR kinase domain, have been proven to be clinically effective in addressing the above problems. The main EGFR-TKIs are summarized in Table 1. In recent decades, EGFR-TKIs have become the first-line standard treatment for patients with advanced NSCLC with EGFR mutations (13,14). The randomized, controlled, large-sample phase-III Iressa Pan-Asia Study (I-PASS) landmark clinical trial was the first to show that gefitinib significantly prolonged progression-free survival (PFS) in the first-line treatment of patients with advanced NSCLC with the EGFR mutation [median PFS: 9.5 vs. 6.3 months; hazard ratio (HR) =0.48, 95% confidence interval (CI): 0.36–0.64; P<0.001], improved the tumor response rate and quality of life (QoL), and had good safety (15). Subsequently, compared with standard chemotherapy, multiple prospective phase-III clinical trials with EGFR-positive patients, such as WJTOG3405 (16), NEJ002 (17), and EURTAC, have shown that the EGFR mutation is an important target and a key predictive marker for first-line EGFR-TKI therapy (18). In addition, the results of the CTONG0806 trial suggest that gefitinib does not significantly improve PFS, tumor response, and QoL in NSCLC patients without EGFR mutations. Thus, EGFR-targeted therapy should not be used in EGFR-negative patients, as it is not beneficial in reducing the risk of disease progression or death (19).

Table 1

Summary of the main EGFR-TKIs

Classification Drug Approval Structure
First-generation EGFR-TKI Gefitinib (Iressa) FDA, EMA, CFDA
Erlotinib (Tarceva) FDA, EMA
Icotinib CFDA
Second-generation EGFR-TKI Afatinib (Gilotrif) FDA, EMA
Third-generation EGFR-TKI Osimertinib (Tagrisso) FDA, EMA, CFDA

EGFR, epidermal growth factor receptor; TKIs, tyrosine kinase inhibitors; FDA, Food and Drug Administration; EMA, European Medicines Agency; CFDA, China Food and Drug Administration.

Unfortunately, after experiencing tumor regression, delayed progression and symptom improvement, research has shown that patients who are treated with EGFR-TKIs inevitably become drug resistant (most often within 9–12 months), which has been defined as “acquired resistance” (20-22). Possible mechanisms for acquired resistance have been extensively discussed; Threonine to Methionine substitution at position 790 (T790M), which is present in approximately 50% of EGFR-TKI resistant NSCLC patients, is the most common alteration (23). Other mechanisms leading to resistance include the human epithelial growth factor receptor 2 (HER2) mutation and amplification (24,25), mesenchymal-epithelial transition (MET) amplification (26), epithelial-mesenchymal transition (EMT) (27), and SCLC phenotypic transformation (28).

Due to the tumor heterogeneity and the possibility of simultaneous or sequential drug resistance mechanisms in patients, combination therapy has been put forward as a promising strategy for the treatment of EGFR-mutated NSCLC. Theoretically, in addition to overcoming or delaying the drug resistance, combination therapy could also enhance anti-cancer efficacy through its synergistic effects (29,30). Additionally, some combination therapy strategies may lower the dose of highly toxic drugs and minimize adverse reactions, and thus expand drug applications (31). In this review, we summarized EGFR-TKI combination strategies for treating EGFR-mutated NSCLC, including combined targeted therapy, radiotherapy, chemotherapy, and immunotherapy, which are of great significance in prolonging the survival of patients with NSCLC. This article is written in accordance with the Narrative Review reporting checklist (available at https://tcr.amegroups.com/article/view/10.21037/tcr-23-956/rc).


Methods

The following Medical Subject Headings (MeSH) terms were used to search the PubMed database: “EGFR-mutated non-small cell lung cancer” and “clinical trial”. Articles with those terms, published in English, from January 1, 2001 to April 15, 2023, were retrieved. Google Scholar was also reviewed to retrieve additional articles. The relevant literature was searched using the following keywords: “EGFR-TKIs”, “combination therapy”, “EGFR-mutated NSCLC”, “acquired resistance”, and “clinical trial”. The research selection process was divided into the following three stages: title review, abstract review and full-text review. Original articles and review articles appropriate to the topic of this review were included in the full-text review phase. The search strategy is detailed in Table 2.

Table 2

The search strategy summary

Items Specification
Date of search February 1 (first search), 2023 to July 15 (last search), 2023
Databases and other sources searched PubMed database, Google Scholar
Search terms used “EGFR-TKIs”, “combination therapy”, “EGFR-mutated NSCLC”, “acquired resistance”, “clinical trial”
Timeframe January 1, 2001 to April 15, 2023
Inclusion and exclusion criteria Inclusion: the search was limited to articles published in English. The research selection process was divided into the following three stages: title review, abstract review, and full-text review. Original articles and review articles appropriate to the topic of this review were included in the full-text review phase
Exclusion: articles not published in English and not related to the research topic were excluded
Selection process Q Zhang conducted the article selection independently. L Xu and R Wang supervised the article selection

EGFR, epidermal growth factor receptor; TKIs, tyrosine kinase inhibitors; NSCLC, non-small cell lung cancer.


EGFR-TKIs combined with targeted therapy

Targeted therapy has been defined as a method of treatment that blocks the growth of cancer cells by interfering with the specific cell molecules required for carcinogenesis and tumor growth, and has fewer side effects than classical cytotoxic chemotherapy (32). In NSCLC patients with EGFR mutations, targeted drugs for different targets can be provided according to different drug resistance mechanisms (see Figure 1) to develop precise and individualized anti-tumor programs.

Figure 1 Various mechanisms of EGFR-TKI resistance. EGF, epidermal growth factor; EGFR, epidermal growth factor receptor; HGF, hepatocyte growth factor; MET, mesenchymal-epithelial transition; HER, human epidermal growth factor receptor; IGF-1, insulin-like growth factor-1; PTEN, phosphatase and tensin homolog deleted on chromosome 10; PI3K, phosphatidylinositol-3 kinase; AKT, protein kinase B; mTOR, mammalian target of rapamycin; JAK, Janus kinase; STAT3, signal transducer and activator of transcription 3; TKI, tyrosine kinase inhibitor.

Targeting MET

The MET receptor, a transmembrane tyrosine kinase receptor encoded by the proto-oncogene MET, is thought to be an important cause of acquired resistance to gefitinib or erlotinib in NSCLC (33). Hepatocyte growth factor (HGF) is a ligand of the MET receptor. It promotes the phosphorylation of MET tyrosine kinase by binding to the MET receptor and then activates the downstream PI3K/AKT/mTOR pathway, which is the key signaling pathway for cell proliferation, survival, and anti-apoptosis (33,34). Thus, targeting MET appears to be an effective approach for treating NSCLC.

Clinical trials have shown exciting results. For example, the INSIGHT study suggests that MET inhibitor tepotinib plus gefitinib results in increased anti-activity compared with standard chemotherapy in patients with EGFR-mutant NSCLC and MET amplification. Unfortunately, the study was prematurely terminated due to insufficient enrollment (35). However, the interim results of this multicenter, open-label, phase-Ib study and the TATTON study suggest that the combination of osimertinib and the MET inhibitor volitinib has an acceptable risk-benefit profile and encouraging anti-tumor activity (36,37). In phase-I studies examining the MET antibody emibetuzumab as a monotherapy or in combination with erlotinib, emibetuzumab showed good tolerance, and no dose-limiting toxicities were observed (38). Based on this study, Camidge et al. conducted a multicenter, randomized, non-controlled, open-label phase-II study, and found that the median PFS and median overall survival (OS) were similar in both the treatment groups, and neither emibetuzumab plus erlotinib nor emibetuzumab monotherapy reversed the acquired resistance to erlotinib in MET positive patients. The study did not meet its primary endpoint; however, a subset of patients obtained meaningful clinical benefits in both treatment groups, including a partial response (PR) lasting up to 11 months (39). It should also be noted that the association of meaningful clinical benefits with the combination treatment strategy has yet to be investigated (39). In addition, exploratory biomarker analyses of other MET-targeted drugs, such as onartuzumab or tivantinib, in combination with erlotinib have shown that favorable trends in PFS were associated with increased MET expression by immunohistochemistry (40,41). Thus, MET remains a valid target in the treatment of EGFR-mutated NSCLC.

Targeting HER2/human epidermal growth factor receptor 3 (HER3)

Other than EGFR, HER2 is one of the most popular targets of NSCLC targeted therapy. HER2 is a transmembrane glycoprotein receptor with intracellular tyrosine kinase activity. Alterations in HER2, including mutation, amplification, and overexpression, have the potential to induce oncogenic effects, and have been detected in many cancers, such as NSCLC (42). Notably, the other target, HER3, cannot be autophosphorylated due to impaired kinase activity but can be tyrosine phosphorylated if it is coupled with other receptor tyrosine kinases (RTKs), such as EGFR and HER2. EGFR and HER2 must recruit and transphosphorylate PI3K through HER3 (43,44). Activated HER3 provides multiple docking sites for PI3K, and amplifies signaling in the PI3K/AKT/mTOR pathway, ultimately promoting cellular survival (45). Thus, HER3 is also considered one of the targets for anti-cancer treatment, especially in EGFR-mutated NSCLC (44).

The results of monotherapies with anti-HER drugs are not optimistic. In a multicenter international trial, trastuzumab (an anti-HER2 drug) was found to lead to a high proportion of remission and provide lasting clinical benefits in patients with advanced HER2-mutated NSCLC; however, the safety of the drug has yet to be studied, and issues such as drug-related interstitial lung disease (ILD) are important risks of the treatment that require careful monitoring and management (46). Additionally, positive results from clinical studies of combination therapies with anti-HER drugs and EGFR-TKIs have rarely been reported. Hughes et al. published the results of a phase-II study exploring the combination of pertuzumab (an anti-HER2/HER3 drug) and erlotinib in 41 patients with relapsed NSCLC. Contrary to the original intention to produce greater activity by targeting the HER family, the combination therapy showed only moderate anti-tumor efficacy and was generally poorly tolerated, which limits its clinical applicability (47). Thus, further research needs to be conducted to discover combinations of drugs that can effectively interfere with HER receptor signaling while demonstrating enhanced tolerability profiles. Further, until powerful, safe and effective drugs targeting HER are developed, treatments in combination with EGFR-TKIs and other target inhibitors, such as mTOR in the downstream pathway, should be preferred.

Targeting PI3K/AKT/mTOR

PI3K-AKT-mTOR is a signaling pathway controlled by the EGFR. PI3K is triggered when extracellular ligands bind to receptors (e.g., when EGF binds to EGFR resulting in the phosphorylation and activation of receptor tyrosine kinase (RTK) to catalyze the phosphorylation of phosphatidylinositol diphosphate into phosphatidylinositol 3-phosphate (PIP3). The accumulation of PIP3 localizes AKT to the plasma membrane, where AKT is phosphorylated by 3-phosphoinositol-dependent kinase 1 indirectly or directly. The activated AKT regulates the phosphorylation of the downstream effectors, instigating alterations in gene expression and cell behavior. The major downstream effectors are mammalian target of rapamycin 1 (mTORC1) and mammalian target of rapamycin complex 2 (mTORC2) (48). Hyperactivation of PI3K/AKT/mTOR signaling not only explains the formation and growth of many tumors, but also illustrates the resistance to targeted inhibitors, such as EGFR-TKIs (49,50). Phosphatase and tensin homolog (PTEN) is a known tumor suppressor gene that can counteract the activation of AKT driven by PI3K. PTEN inactivation is associated with resistance to EGFR-TKI treatment and lower survival in NSCLC patients (50).

Buparlisib (BKM120) is an oral pan-class I, reversible inhibitor of PI3K. Tan et al. observed mild anti-tumor activity with gefitinib and buparlisib in patients resistant to EGFR-TKIs. However, given the late toxicity and long half-life period of buparlisib, other alternatives need to be explored, such as copanlisib (BAY80-6946), duvelisib (IPI-145), and pictilisib (GDC-0941) (51). In addition, there are dual PI3K/mTOR inhibitors, such as dactolisib (NVP-BEZ235), and apitolisib (GDC-0980). Despite a reasonable theoretical basis and demonstrable pharmacodynamic tumor activity in the relevant tumor population, it is not possible to determine a dose and schedule for this class of drugs that is both tolerated and provides clear efficacy in the population evaluated. Thus, their combination with EGFR-TKIs is still in the early clinical development stage (52).

Molife et al. and Lin et al. each conducted a phase-I trial evaluating the oral AKT inhibitor MK-2206 plus erlotinib or gefitinib, and both reported that they were well tolerated and found early evidence of anti-tumor activity (53,54). Based on these promising results, Lara et al. conducted a phase-II study and found that the combination of MK-2206 plus erlotinib was more effective in patients with EGFR wild-type NSCLC than in patients with EGFR-mutant NSCLC. Some efficacy was observed in the EGFR-mutated NSCLC; however, this did not exceed the previous estimates. Thus, AKT pathway inhibition merits additional clinical assessment in EGFR wild-type NSCLC (55). In general, there is limited literature on the combination of AKT inhibitors and EGFR-TKIs in the treatment of NSCLC in recent years, and relevant clinical studies are not yet mature.

Rapamycin and its analogues are currently the inhibitors most commonly used to target the PI3K/AKT/mTOR pathway. Among them, everolimus (RAD001), an orally bioavailable derivative of rapamycin, is a potent mTOR inhibitor. It has been shown to be highly effective in restoring the sensitivity of gefitinib-resistant NSCLC cell lines with the PIK3CA mutation or PTEN deletion (56,57). Many clinical trials have been conducted to assess the efficacy of combining everolimus with EGFR-TKIs. Compared with promising pre-clinical trial results, the efficacy of this combination in patients with EGFR-mutated NSCLC appears to be unsatisfactory (58). In a multicenter, open-label, phase-II study, 133 patients received everolimus-erlotinib (n=66) or erlotinib alone (n=67), and at 3 months, had disease control rate (DCR) of 39.4% and 28.4%, respectively. It was estimated that the probability that the difference in the DCR at 3 months would be ≥15% was 29.8%, which was below the prespecified probability threshold of ≥40%. The median PFS for the patients who received everolimus-erlotinib (n=66) and those who received erlotinib alone was 2.9 and 2.0 months, respectively. Grade 3/4 adverse events occurred in 72.7% and 32.3% of these patients, respectively. Thus, everolimus plus erlotinib was not considered effective enough, and the combination does not warrant further investigation due to the increased toxicity and the lack of substantial improvement in disease stabilization (59). Similarly, a phase-II study by Price et al., which achieved a partial response (PR) in eight of 62 patients and a response rate of only 13%, did not meet the prespecified response threshold to warrant further investigations of everolimus in combination with gefitinib (60). In addition, Fang et al. also found that everolimus, whether combined with gefitinib, afatinib, or osimertinib, showed very limited anti-tumor activity in EGFR-TKI-resistant NSCLC patients (61).

Targeting JAK2/signal transducer and activator of transcription 3 (STAT3)

Ligand-binding receptors recruit and phosphorylate JAK2 (a member of the JAK family that also includes JAK1, JAK3, and Tyk2, among which, JAK2 plays a prominent role in tumorigenesis), which ultimately leads to STAT3 protein phosphorylation, dimerization, and activation (62). It should be noted that in addition to JAK, STAT3 activation can also be mediated by the Src family, Abl family, EGFR, and insulin-like growth factor-1 receptor (IGF-1R) (63). After STAT3 is activated by JAK2, it is transported into the nucleus through the nuclear membrane, regulates the expression of related genes, and participates in important biological processes, such as cell proliferation, differentiation, apoptosis, and angiogenesis (62,64). Similar to PI3K/AKT/mTOR, JAK2/STAT3 also has a significant role in tumorigenesis, and its abnormal activation is also involved in the decreased sensitivity of NSCLC to EGFR-TKIs (65).

The phase-Ib study by Park et al. demonstrated that the combination of afatinib and the JAK1/2 inhibitor ruxolitinib was tolerated by patients, with moderate clinical activity observed in NSCLC patients with acquired resistance to EGFR-TKIs. The 30 patients had an objective response rate (ORR) of 23.3%, a DCR of 93.3% (no patients achieved a complete response, seven achieved a PR, and 21 achieved stable diseases), and a median PFS of 4.9 (95% CI: 2.4–7.5) months (66). However, another phase-Ib study concluded that JAK1/2 inhibitor momelotinib in combination with erlotinib did not appear to produce any improved benefits compared to findings based on historical data of erlotinib monotherapy in patients diagnosed with EGFR-mutated NSCLC (67). Thus, the efficacy of different combinations of JAK inhibitors and EGFR-TKIs needs to be further explored in clinical studies.

Research is still being conducted on the safety and anti-tumor efficacy of STAT3 inhibitors as monotherapies. For example, the STAT3 inhibitor OPB-51602 has been shown to display significant anti-tumor activity, especially in NSCLC; however, its long half-life period and the poorer tolerability of continuous dosing, compared with intermittent dosing, suggests that less frequent dosing should be explored (68). Zheng et al. also found that the STAT3 inhibitor W2014-S showed significant anti-tumor activity in NSCLC (69). This suggests that combinations of STAT3 inhibitors and EGFR-TKIs may be a potential strategy for overcoming EGFR-TKI-acquired resistance in NSCLC patients.

Targeting IGF-1R

IGF-1R is a transmembrane heterotetramer comprising two extracellular α and β subunits (70). Upon the binding of IGF-1 and IGF-2 to the extracellular subunit domain of IGF-1R, the tyrosine kinase activity of IGF-1R is activated, and the activation of IGF-1R initiates cascades involving signal transduction pathways, such as Ras, Raf, and MAPK (71). The overexpression of IGF-1R has been reported to promote tumor growth, progression, invasion, and metastasis (72). IGF-1R also plays an important role in promoting the progression of NSCLC (73).

Many pre-clinical studies have shown that the joint administration of IGF-1R inhibitors, such as α-IR3, AG1024, and R1507, with EGFR-TKIs amplifies the suppressive effects on growth and apoptosis initiated by EGFR-TKIs, which could provide a potential way of treating EGFR-TKI-resistant NSCLC (74,75). However, very few relevant clinical studies have been conducted in this area.

Targeting rearranged during transfection (RET)

The RET proto-oncogene encodes a transmembrane RTK involved in normal embryonic development (76). Due to an aberrant DNA repair process, RET fuses with another irrelevant gene (77), activating various downstream signaling cascades that play essential roles in cell proliferation and survival; that is, the PI3K/AKT, JAK2/STAT3 pathways (78). It has emerged as a rare but targetable acquired resistance mechanism in EGFR-mutated NSCLC patients undergoing treatment with EGFR-TKI (79,80).

Thus, a combination therapy of anti-EGFR and anti-RET therapy will likely be required to overcome this resistance. Notably, two highly potent RET inhibitors, selpercatinib and pralsetinib, have been proven to be effective in treating advanced or metastatic RET-altered NSCLC (80). Additionally, RET fusions are more likely to be associated with EGFR-mutant NSCLC patients who received therapeutic interventions targeting EGFR through third-generation EGFR-TKIs (80). In a multicenter, prospectively treated cohort, Rotow et al. found that the addition of selpercatinib to osimertinib was feasible and safe and provided clinical benefits to patients with EGFR-mutant NSCLC with an acquired RET fusion (81). Additionally, Piotrowska et al. reported that RET fusions mediate resistance to EGFR inhibitors and demonstrated that combined EGFR and RET inhibition with osimertinib/pralsetinib (BLU-667) may be a well-tolerated and effective treatment strategy for EGFR-mutant NSCLC (82). Urbanska et al. also found that EGFR-mutated patients displayed sustained ongoing objective response (OR) to an osimertinib-pralsetinib combination for more than 12 months, which provides clinical evidence of the effectively targetable mechanism of osimertinib resistance (83).


EGFR-TKIs combined with chemotherapy

As the first-line treatment for NSCLC patients without clear therapeutic targets, chemotherapy has always played an important role in tumor treatments (84). Platinum-based therapy is the mainstay of chemotherapy for NSCLC patients and is usually administered in combination with a tubulin binding agent [such as taxanes (paclitaxel or docetaxel) and vinca alkaloids (vinorelbine or vincristine)], a camptothecin analogue (irinotecan or opotecan), gemcitabine, and pemetrexed (85). It should be noted that pemetrexed has significant activity, favorable tolerance, and low toxicity compared to other cytotoxic agents. Extensive research has been conducted to explore its administration either alone or in combination with other regimes (86). To ameliorate the inevitable drug-resistant outcome of EGFR-TKI therapy, combining it with chemotherapy could provide an effective treatment option for EGFR-mutated NSCLC patients.

Studies of the combination of EGFR-TKIs and chemotherapy in patients with EGFR mutations have shown promising results. The NEJ009 study showed that patients with advanced NSCLC with EGFR mutations treated with gefitinib combined with carboplatin plus pemetrexed had better PFS (20.9 vs. 11.2 months, respectively, HR =0.493, P<0.001) with an acceptable toxicity profile than those treated with gefitinib alone. The benefits of this combined treatment in terms of OS require further validation (but the results of the study showed that the patients had an OS of 52.2 vs. 38.8 months, respectively, HR =0.695, P=0.0013) (87). Additionally, Planchard et al. and Watanabe et al. found that at the recommended doses, osimertinib or afatinib in combination with carboplatin and pemetrexed also had manageable safety and tolerability, and good clinical efficacy in patients with EGFR-mutated NSCLC (88,89). A meta-analysis of 18 eligible trials involving 4,628 patients and 12 treatments indicated that osimertinib and gefitinib plus pemetrexed-based chemotherapy were associated with the best PFS and OS benefits for patients with advanced EGFR-mutated NSCLC compared with other first-line treatments (90). Another meta-analysis of eight randomized controlled trials involving 1,349 advanced NSCLC patients with the sensitive EGFR mutation had similar findings, which suggests that compared with EGFR-TKI monotherapy, the combination of first-generation EGFR-TKI and chemotherapy, especially when applying the concurrent delivery of platinum-based doublet chemotherapeutic drugs, significantly improves the ORR, PFS, and OS in the first-line treatment of advanced EGFR-mutated NSCLC. Despite the increased incidence of chemotherapy-induced toxicities in the combination group, it is well tolerated and can be effectively managed from a clinical perspective (91). However, Gijtenbeek et al. demonstrated that the effects of erlotinib with cisplatin-pemetrexed were not favorable, as the toxicity rate was high and not negligible (92). Moreover, another systematic analysis reported that the combination of chemotherapy and EGFR-TKIs did not achieve satisfactory results, and that while there was no notable difference in OS and the ORR, there was an increased incidence of grade 3/4 anemia, rash, and other adverse events in the patients (93). As the role of EGFR-TKIs combined with chemotherapy remains controversial, it has not yet been widely used in clinical practice, and further clinical trials need to be conducted to explore and verify its use (31).


EGFR-TKIs combined with radiotherapy

Radiotherapy is an important way of treating NSCLC, and its role in locally advanced and early-stage disease, recurrence, postoperative, central early stage NSCLC, and multiple primary lung cancer has been fully demonstrated (94). Several studies have been carried out to test the hypothesis that the combination of EGFR-TKIs and radiotherapy may confer better survival benefits than monotherapy.

A retrospective study of 380 patients reported that the EGFR-TKI + thoracic stereotactic body radiation therapy (SBRT) group had a median PFS of 19.4 months and the EGFR-TKI group had a median PFS of 13.7 months (P=0.034) with tolerable toxicity and no significant effect in relation to OS (P=0.557) (95). The clinical trials by Zheng et al. and Akamatsu et al. also validated the efficacy and tolerability of erlotinib or gefitinib plus thoracic radiotherapy in a subset of NSCLC patients harboring EGFR mutations (96,97). Notably, an especially high incidence of grade two or worse radiation pneumonia was observed in patients receiving osimertinib combined with thoracic radiotherapy (63.6%) compared with those receiving erlotinib or gefitinib combined with thoracic radiotherapy, which should serve as a warning to physicians to exercise caution when using this combination regimen (98).

In addition to thorax, between 25% and 40% of NSCLC patients reportedly develop brain metastases during the course of the disease, usually within 2 years of diagnosis of the primary tumor and have poor prospects (99). Current treatments for patients with brain metastases include whole-brain radiation therapy (WBRT) with or without stereotactic radiosurgery (100). The blood-brain barrier renders most chemotherapeutic agents ineffective, but it has been established that EGFR-TKIs can permeate the blood-brain barrier (101) with limited penetration into cerebrospinal fluid (102,103), and thus could be administered with WBRT.

Many studies have reported on the feasibility of WBRT combined with EFGR-TKIs in the treatment of NSCLC patients with the EGFR mutation and brain metastases. Fan et al. reported a median survival time of 22.0 months for patients with EGFR mutations and 7.5 months for those with wild-type EGFR (P=0.0001), which suggests that patients with EGFR mutations benefit more from icotinib combined with WBRT (104). Additionally, a meta-analysis of 18 prospective clinical studies reported that the ORR of WBRT combined with erlotinib/gefitinib was also superior to that of WBRT alone (odds ratio =2.67; 95% CI: 2.10–3.38; P<0.05) (105). Moreover, He et al. found that in first-line therapy for NSCLC patients with EGFR mutations, the combination therapy of EGFR-TKIs and WBRT significantly improved OS and intracranial PFS (iPFS), and patients with symptomatic brain metastasis, an older age, and exon 19 deletion may benefit more from combination therapy (106). However, He et al. also suggested that EGFR-TKI alone may be an option as a first-line therapy for patients with three or less brain metastases, which may defer or avoid the neurocognitive sequelae caused by WBRT (106). In addition, one case report demonstrated that the combination of WBRT and afatinib might cause serious dermatological toxicity (107). Thus, more multi-institutional, prospective randomized clinical trials need to be conducted to further explore the combination of EGFR-TKIs and radiotherapy to better guide clinical treatment.


EGFR-TKIs combined with immunotherapy

Immunotherapy has become popular as the preferred option for cancer treatment in recent years (108). It aims to improve the anti-tumor immune response. Related drugs stimulate or promote the activation of the immune system to kill tumor cells that have escaped previous immunological surveillance. Compared to chemotherapy and other drugs that kill cancer cells directly, immunotherapy has fewer off-target effects (108). Thus, immunotherapy exerts a powerful anti-cancer effect by improving the immune microenvironment and has been approved for the treatment of NSCLC (109). The combination of immunotherapy with EGFR-TKIs is also under evaluation.

The programmed cell death protein-1 (PD-1) and its key ligand, the programmed cell death ligand-1 (PD-L1), are key therapeutic targets for NSCLC therapy. PD-L1 induces T cell apoptosis by binding to its receptor PD-1, which is mainly expressed in activated T cells (110,111). The overexpression of PD-L1 is associated with a poor prognosis in many cancers, such as NSCLC and breast cancer (112). A phase-I open-label multicenter study (NCT02088112) observed encouraging activity in durvalumab (an anti-PD-L1 antibody) and gefitinib in NSCLC patients with sensitizing EGFR mutations (113). The TATTON study is a multi-arm, open-label, phase-Ib study designed to evaluate the safety and tolerability of osimertinib based combinations (osimertinib + MEK1-2 inhibitor selumetinib, osimertinib + MET-TKI savolitinib, and osimertinib + anti-PD-L1 monoclonal antibody durvalumab) in patients with EGFR-mutated NSCLC. As part of the TATTON study, Oxnard et al. found that while osimertinib (80 mg orally once a day) combined with durvalumab (3–10 mg/kg intravenously every 2 weeks) was tolerable, there was a higher than expected frequency of ILD (22%). However, the mechanism underlying the high incidence of ILD remains unclear, which led to the discontinuation of this combination (114).

Cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) is another immune checkpoint receptor that induces T cell non-reactivity by binding to the B7 molecule and participates in the negative regulation of the immune response (115). It has also been developed as a prospective target for therapy in cases of NSCLC (115). In a small phase-I trial, Chalmers et al. evaluated a combination of ipilimumab (anti-CTLA-4 antibody) plus erlotinib in 11 NSCLC patients with EGFR mutations. This combination was associated with excessive short-term gastrointestinal toxicity (36%), which exceeded the pre-planned conventional definition of an unacceptable toxicity frequency of 33%, leading to the early termination of the study. However, the long-term follow-up revealed an unexpected prolongation of patient survival, with a median PFS of 27.8 months and a median OS of 42.3 months (116). Based on the study by Chalmers et al., Puri et al. replaced erlotinib with osimertinib to decrease the toxicity of the combination therapy to assess the efficacy of the combination of ipilimumab and osimertinib in patients with EGFR-mutated NSCLC. The study is ongoing and the results are expected to be published in the near future (117).

In addition, high levels of circulating vascular endothelial growth factor (VEGF) stimulate tumor angiogenesis, which plays an important role in the growth, proliferation, and metastasis of tumor cells in NSCLC patients (118). ARTEMIS-CTONG1509, a multicenter phase-III study, found that bevacizumab (an anti-VEGF antibody) plus erlotinib significantly improved PFS in patients with EGFR-mutated NSCLC, including those with brain metastases at the baseline (119). Kuo et al. found that a bevacizumab combination treatment showed moderate efficacy in afatinib-treated NSCLC patients with the EGFR-sensitizing mutation (120). However, a treatment of osimertinib plus bevacizumab failed to show any efficacy in improving the PFS of EGFR-mutated NSCLC patients, with similar findings reported in the studies of Soo et al. and Kenmotsu et al. (121,122).

As mentioned above, most studies have shown that a combination of immunotherapy drugs and EGFR-TKIs could be comparatively clinically effective. However, EGFR-TKIs can cause several serious adverse events, notably including interstitial pneumonia. Given the potential toxicity challenges associated with combination therapies, some studies have had to be terminated prematurely. Widespread safety concerns indicate a lack of understanding of antibody-based immunotherapy and should be addressed before the extensive clinical use of immunotherapy drugs.


Conclusions

In summary, the use of EGFR-TKI combination therapy is case-dependent. Most clinical trials have shown the efficacy and safety of EGFR-TKI combination therapy in patients with EGFR-mutated NSCLC; however, in some instances, several serious adverse events have led to the early termination of trials. Thus, more clinical studies with large sample sizes need to be conducted to analyze the activity and toxicity of combination therapies to explore potential and well-tolerated options. In addition, retrospective studies should be carried out from which the classification of patient subgroups should be analyzed to select patients who might benefit from combination therapy in terms of cost-effectiveness, increased longevity, and improved QoL. In conclusion, EGFR-TKI combination therapy is an effective approach for the treatment of patients with EGFR-mutated NSCLC and deserves further development.


Acknowledgments

Funding: None.


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://tcr.amegroups.com/article/view/10.21037/tcr-23-956/rc

Peer Review File: Available at https://tcr.amegroups.com/article/view/10.21037/tcr-23-956/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tcr.amegroups.com/article/view/10.21037/tcr-23-956/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Bade BC, Dela Cruz CS. Lung Cancer 2020: Epidemiology, Etiology, and Prevention. Clin Chest Med 2020;41:1-24. [Crossref] [PubMed]
  2. Chhikara BS, Parang K. Global Cancer Statistics 2022: the trends projection analysis. Chemical Biology Letters 2023;10:451.
  3. Gelatti ACZ, Drilon A, Santini FC. Optimizing the sequencing of tyrosine kinase inhibitors (TKIs) in epidermal growth factor receptor (EGFR) mutation-positive non-small cell lung cancer (NSCLC). Lung Cancer 2019;137:113-22. [Crossref] [PubMed]
  4. Zarogoulidis K, Zarogoulidis P, Darwiche K, et al. Treatment of non-small cell lung cancer (NSCLC). J Thorac Dis 2013;5:S389-96. [PubMed]
  5. O'Leary C, Gasper H, Sahin KB, et al. Epidermal Growth Factor Receptor (EGFR)-Mutated Non-Small-Cell Lung Cancer (NSCLC). Pharmaceuticals (Basel) 2020;13:273. [Crossref] [PubMed]
  6. Butti R, Das S, Gunasekaran VP, et al. Receptor tyrosine kinases (RTKs) in breast cancer: signaling, therapeutic implications and challenges. Mol Cancer 2018;17:34. [Crossref] [PubMed]
  7. Evangelopoulos ME, Weis J, Krüttgen A. Signalling pathways leading to neuroblastoma differentiation after serum withdrawal: HDL blocks neuroblastoma differentiation by inhibition of EGFR. Oncogene 2005;24:3309-18. [Crossref] [PubMed]
  8. Liao BC, Lee JH, Lin CC, et al. Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors for Non-Small-Cell Lung Cancer Patients with Leptomeningeal Carcinomatosis. J Thorac Oncol 2015;10:1754-61. [Crossref] [PubMed]
  9. Nicholson RI, Gee JM, Harper ME. EGFR and cancer prognosis. Eur J Cancer 2001;37:S9-15. [Crossref] [PubMed]
  10. Zhu H, Acquaviva J, Ramachandran P, et al. Oncogenic EGFR signaling cooperates with loss of tumor suppressor gene functions in gliomagenesis. Proc Natl Acad Sci U S A 2009;106:2712-6. [Crossref] [PubMed]
  11. Purba ER, Saita EI, Maruyama IN. Activation of the EGF Receptor by Ligand Binding and Oncogenic Mutations: The "Rotation Model". Cells 2017;6:13. [Crossref] [PubMed]
  12. Scaltriti M, Baselga J. The epidermal growth factor receptor pathway: a model for targeted therapy. Clin Cancer Res 2006;12:5268-72. [Crossref] [PubMed]
  13. Paez JG, Jänne PA, Lee JC, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 2004;304:1497-500. [Crossref] [PubMed]
  14. Pao W, Chmielecki J. Rational, biologically based treatment of EGFR-mutant non-small-cell lung cancer. Nat Rev Cancer 2010;10:760-74. [Crossref] [PubMed]
  15. Fukuoka M, Wu YL, Thongprasert S, et al. Biomarker analyses and final overall survival results from a phase III, randomized, open-label, first-line study of gefitinib versus carboplatin/paclitaxel in clinically selected patients with advanced non-small-cell lung cancer in Asia (IPASS). J Clin Oncol 2011;29:2866-74. [Crossref] [PubMed]
  16. Mitsudomi T, Morita S, Yatabe Y, et al. Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol 2010;11:121-8. [Crossref] [PubMed]
  17. Maemondo M, Inoue A, Kobayashi K, et al. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med 2010;362:2380-8. [Crossref] [PubMed]
  18. Rosell R, Carcereny E, Gervais R, et al. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol 2012;13:239-46. [Crossref] [PubMed]
  19. Zhou Q, Cheng Y, Yang JJ, et al. Pemetrexed versus gefitinib as a second-line treatment in advanced nonsquamous nonsmall-cell lung cancer patients harboring wild-type EGFR (CTONG0806): a multicenter randomized trial. Ann Oncol 2014;25:2385-91. [Crossref] [PubMed]
  20. Jackman D, Pao W, Riely GJ, et al. Clinical definition of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer. J Clin Oncol 2010;28:357-60. [Crossref] [PubMed]
  21. Leonetti A, Sharma S, Minari R, et al. Resistance mechanisms to osimertinib in EGFR-mutated non-small cell lung cancer. Br J Cancer 2019;121:725-37. [Crossref] [PubMed]
  22. Piper-Vallillo AJ, Sequist LV, Piotrowska Z. Emerging Treatment Paradigms for EGFR-Mutant Lung Cancers Progressing on Osimertinib: A Review. J Clin Oncol 2020; Epub ahead of print. [Crossref] [PubMed]
  23. Papadimitrakopoulou VA, Mok TS, Han JY, et al. Osimertinib versus platinum-pemetrexed for patients with EGFR T790M advanced NSCLC and progression on a prior EGFR-tyrosine kinase inhibitor: AURA3 overall survival analysis. Ann Oncol 2020;31:1536-44. [Crossref] [PubMed]
  24. Wang SE, Narasanna A, Perez-Torres M, et al. HER2 kinase domain mutation results in constitutive phosphorylation and activation of HER2 and EGFR and resistance to EGFR tyrosine kinase inhibitors. Cancer Cell 2006;10:25-38. [Crossref] [PubMed]
  25. Takezawa K, Pirazzoli V, Arcila ME, et al. HER2 amplification: a potential mechanism of acquired resistance to EGFR inhibition in EGFR-mutant lung cancers that lack the second-site EGFRT790M mutation. Cancer Discov 2012;2:922-33. [Crossref] [PubMed]
  26. Cappuzzo F, Marchetti A, Skokan M, et al. Increased MET gene copy number negatively affects survival of surgically resected non-small-cell lung cancer patients. J Clin Oncol 2009;27:1667-74. [Crossref] [PubMed]
  27. Thomson S, Buck E, Petti F, et al. Epithelial to mesenchymal transition is a determinant of sensitivity of non-small-cell lung carcinoma cell lines and xenografts to epidermal growth factor receptor inhibition. Cancer Res 2005;65:9455-62. [Crossref] [PubMed]
  28. Sequist LV, Waltman BA, Dias-Santagata D, et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci Transl Med 2011;3:75ra26. [Crossref] [PubMed]
  29. Liu Z, Gao W. Synergistic effects of Bcl-2 inhibitors with AZD9291 on overcoming the acquired resistance of AZD9291 in H1975 cells. Arch Toxicol 2020;94:3125-36. [Crossref] [PubMed]
  30. Ao L, Fang S, Zhang K, et al. Sequence-dependent synergistic effect of aumolertinib-pemetrexed combined therapy on EGFR-mutant non-small-cell lung carcinoma with pre-clinical and clinical evidence. J Exp Clin Cancer Res 2022;41:163. [Crossref] [PubMed]
  31. Rebuzzi SE, Alfieri R, La Monica S, et al. Combination of EGFR-TKIs and chemotherapy in advanced EGFR mutated NSCLC: Review of the literature and future perspectives. Crit Rev Oncol Hematol 2020;146:102820. [Crossref] [PubMed]
  32. Joo WD, Visintin I, Mor G. Targeted cancer therapy--are the days of systemic chemotherapy numbered? Maturitas 2013;76:308-14. [Crossref] [PubMed]
  33. Appleman LJ. MET signaling pathway: a rational target for cancer therapy. J Clin Oncol 2011;29:4837-8. [Crossref] [PubMed]
  34. Nakamura T, Sakai K, Nakamura T, et al. Hepatocyte growth factor twenty years on: Much more than a growth factor. J Gastroenterol Hepatol 2011;26:188-202. [Crossref] [PubMed]
  35. Wu YL, Cheng Y, Zhou J, et al. Tepotinib plus gefitinib in patients with EGFR-mutant non-small-cell lung cancer with MET overexpression or MET amplification and acquired resistance to previous EGFR inhibitor (INSIGHT study): an open-label, phase 1b/2, multicentre, randomised trial. Lancet Respir Med 2020;8:1132-43. [Crossref] [PubMed]
  36. Sequist LV, Han JY, Ahn MJ, et al. Osimertinib plus savolitinib in patients with EGFR mutation-positive, MET-amplified, non-small-cell lung cancer after progression on EGFR tyrosine kinase inhibitors: interim results from a multicentre, open-label, phase 1b study. Lancet Oncol 2020;21:373-86. [Crossref] [PubMed]
  37. Hartmaier RJ, Markovets AA, Ahn MJ, et al. Osimertinib + Savolitinib to Overcome Acquired MET-Mediated Resistance in Epidermal Growth Factor Receptor-Mutated, MET-Amplified Non-Small Cell Lung Cancer: TATTON. Cancer Discov 2023;13:98-113. [Crossref] [PubMed]
  38. Rosen LS, Goldman JW, Algazi AP, et al. A First-in-Human Phase I Study of a Bivalent MET Antibody, Emibetuzumab (LY2875358), as Monotherapy and in Combination with Erlotinib in Advanced Cancer. Clin Cancer Res 2017;23:1910-9. [Crossref] [PubMed]
  39. Camidge DR, Moran T, Demedts I, et al. A Randomized, Open-Label Phase II Study Evaluating Emibetuzumab Plus Erlotinib and Emibetuzumab Monotherapy in MET Immunohistochemistry Positive NSCLC Patients with Acquired Resistance to Erlotinib. Clin Lung Cancer 2022;23:300-10. [Crossref] [PubMed]
  40. Scagliotti G, von Pawel J, Novello S, et al. Phase III Multinational, Randomized, Double-Blind, Placebo-Controlled Study of Tivantinib (ARQ 197) Plus Erlotinib Versus Erlotinib Alone in Previously Treated Patients With Locally Advanced or Metastatic Nonsquamous Non-Small-Cell Lung Cancer. J Clin Oncol 2015;33:2667-74. [Crossref] [PubMed]
  41. Spigel DR, Edelman MJ, O'Byrne K, et al. Results From the Phase III Randomized Trial of Onartuzumab Plus Erlotinib Versus Erlotinib in Previously Treated Stage IIIB or IV Non-Small-Cell Lung Cancer: METLung. J Clin Oncol 2017;35:412-20. [Crossref] [PubMed]
  42. Ren S, Wang J, Ying J, et al. Consensus for HER2 alterations testing in non-small-cell lung cancer. ESMO Open 2022;7:100395. [Crossref] [PubMed]
  43. Yonesaka K, Tanizaki J, Maenishi O, et al. HER3 Augmentation via Blockade of EGFR/AKT Signaling Enhances Anticancer Activity of HER3-Targeting Patritumab Deruxtecan in EGFR-Mutated Non-Small Cell Lung Cancer. Clin Cancer Res 2022;28:390-403. [Crossref] [PubMed]
  44. Kawakami H, Yonesaka K. HER3 and its Ligand, Heregulin, as Targets for Cancer Therapy. Recent Pat Anticancer Drug Discov 2016;11:267-74. [Crossref] [PubMed]
  45. Engelman JA, Jänne PA, Mermel C, et al. ErbB-3 mediates phosphoinositide 3-kinase activity in gefitinib-sensitive non-small cell lung cancer cell lines. Proc Natl Acad Sci U S A 2005;102:3788-93. [Crossref] [PubMed]
  46. Li BT, Smit EF, Goto Y, et al. Trastuzumab Deruxtecan in HER2-Mutant Non-Small-Cell Lung Cancer. N Engl J Med 2022;386:241-51. [Crossref] [PubMed]
  47. Hughes B, Mileshkin L, Townley P, et al. Pertuzumab and erlotinib in patients with relapsed non-small cell lung cancer: a phase II study using 18F-fluorodeoxyglucose positron emission tomography/computed tomography imaging. Oncologist 2014;19:175-6. [Crossref] [PubMed]
  48. Gadgeel SM, Wozniak A. Preclinical rationale for PI3K/Akt/mTOR pathway inhibitors as therapy for epidermal growth factor receptor inhibitor-resistant non-small-cell lung cancer. Clin Lung Cancer 2013;14:322-32. [Crossref] [PubMed]
  49. Yu L, Wei J, Liu P. Attacking the PI3K/Akt/mTOR signaling pathway for targeted therapeutic treatment in human cancer. Semin Cancer Biol 2022;85:69-94. [Crossref] [PubMed]
  50. Pérez-Ramírez C, Cañadas-Garre M, Molina MÁ, et al. PTEN and PI3K/AKT in non-small-cell lung cancer. Pharmacogenomics 2015;16:1843-62. [Crossref] [PubMed]
  51. Tan DSW, Lim KH, Tai WM, et al. A phase Ib safety and tolerability study of a pan class I PI3K inhibitor buparlisib (BKM120) and gefitinib (gef) in EGFR TKI-resistant NSCLC. J Clin Oncol 2013;31:abstr 8107.
  52. Yang J, Nie J, Ma X, et al. Targeting PI3K in cancer: mechanisms and advances in clinical trials. Mol Cancer 2019;18:26. [Crossref] [PubMed]
  53. Molife LR, Yan L, Vitfell-Rasmussen J, et al. Phase 1 trial of the oral AKT inhibitor MK-2206 plus carboplatin/paclitaxel, docetaxel, or erlotinib in patients with advanced solid tumors. J Hematol Oncol 2014;7:1. [Crossref] [PubMed]
  54. Lin CC, Yu CJ, Ho CC, et al. A Phase I Dose Defining Study for MK-2206 Combined with Gefitinib in NSCLC Population Enriched with EGFR Mutation. Ann Oncol 2015;26:abstr II16.
  55. Lara PN Jr, Longmate J, Mack PC, et al. Phase II Study of the AKT Inhibitor MK-2206 plus Erlotinib in Patients with Advanced Non-Small Cell Lung Cancer Who Previously Progressed on Erlotinib. Clin Cancer Res 2015;21:4321-6. [Crossref] [PubMed]
  56. Dong S, Zhang XC, Cheng H, et al. Everolimus synergizes with gefitinib in non-small-cell lung cancer cell lines resistant to epidermal growth factor receptor tyrosine kinase inhibitors. Cancer Chemother Pharmacol 2012;70:707-16. [Crossref] [PubMed]
  57. La Monica S, Galetti M, Alfieri RR, et al. Everolimus restores gefitinib sensitivity in resistant non-small cell lung cancer cell lines. Biochem Pharmacol 2009;78:460-8. [Crossref] [PubMed]
  58. Fang W, Huang Y, Gu W, et al. PI3K-AKT-mTOR pathway alterations in advanced NSCLC patients after progression on EGFR-TKI and clinical response to EGFR-TKI plus everolimus combination therapy. Transl Lung Cancer Res 2020;9:1258-67. [Crossref] [PubMed]
  59. Besse B, Leighl N, Bennouna J, et al. Phase II study of everolimus-erlotinib in previously treated patients with advanced non-small-cell lung cancer. Ann Oncol 2014;25:409-15. [Crossref] [PubMed]
  60. Price KA, Azzoli CG, Krug LM, et al. Phase II trial of gefitinib and everolimus in advanced non-small cell lung cancer. J Thorac Oncol 2010;5:1623-9. [Crossref] [PubMed]
  61. Fang W, Huang Y, Gan J, et al. The impact of PIK3CA/PTEN/AKT1 genes in advanced NSCLC patients with acquired EGFR-TKI resistance and clinical response to EGFR-TKI plus everolimus combination therapy. Cancer Res 2019;79:327. [Crossref]
  62. Xin P, Xu X, Deng C, et al. The role of JAK/STAT signaling pathway and its inhibitors in diseases. Int Immunopharmacol 2020;80:106210. [Crossref] [PubMed]
  63. Chai EZ, Shanmugam MK, Arfuso F, et al. Targeting transcription factor STAT3 for cancer prevention and therapy. Pharmacol Ther 2016;162:86-97. [Crossref] [PubMed]
  64. Bolli R, Dawn B, Xuan YT. Role of the JAK-STAT pathway in protection against myocardial ischemia/reperfusion injury. Trends Cardiovasc Med 2003;13:72-9. [Crossref] [PubMed]
  65. Harada D, Takigawa N, Ochi N, et al. JAK2-related pathway induces acquired erlotinib resistance in lung cancer cells harboring an epidermal growth factor receptor-activating mutation. Cancer Sci 2012;103:1795-802. [Crossref] [PubMed]
  66. Park JS, Hong MH, Chun YJ, et al. A phase Ib study of the combination of afatinib and ruxolitinib in EGFR mutant NSCLC with progression on EGFR-TKIs. Lung Cancer 2019;134:46-51. [Crossref] [PubMed]
  67. Padda SK, Reckamp KL, Koczywas M, et al. A phase 1b study of erlotinib and momelotinib for the treatment of EGFR-mutated, tyrosine kinase inhibitor-naive metastatic non-small cell lung cancer. Cancer Chemother Pharmacol 2022;89:105-15. [Crossref] [PubMed]
  68. Wong AL, Soo RA, Tan DS, et al. Phase I and biomarker study of OPB-51602, a novel signal transducer and activator of transcription (STAT) 3 inhibitor, in patients with refractory solid malignancies. Ann Oncol 2015;26:998-1005. [Crossref] [PubMed]
  69. Zheng Q, Dong H, Mo J, et al. A novel STAT3 inhibitor W2014-S regresses human non-small cell lung cancer xenografts and sensitizes EGFR-TKI acquired resistance. Theranostics 2021;11:824-40. [Crossref] [PubMed]
  70. Boone DN, Lee AV. Targeting the insulin-like growth factor receptor: developing biomarkers from gene expression profiling. Crit Rev Oncog 2012;17:161-73. [Crossref] [PubMed]
  71. Yuan J, Yin Z, Tao K, et al. Function of insulin-like growth factor 1 receptor in cancer resistance to chemotherapy. Oncol Lett 2018;15:41-7. [PubMed]
  72. Yamamoto T, Oshima T, Yoshihara K, et al. Clinical significance of immunohistochemical expression of insulin-like growth factor-1 receptor and matrix metalloproteinase-7 in resected non-small cell lung cancer. Exp Ther Med 2012;3:797-802. [Crossref] [PubMed]
  73. Zhao J, Shi X, Wang T, et al. The Prognostic and Clinicopathological Significance of IGF-1R in NSCLC: a Meta-Analysis. Cell Physiol Biochem 2017;43:697-704. [Crossref] [PubMed]
  74. Choi YJ, Rho JK, Jeon BS, et al. Combined inhibition of IGFR enhances the effects of gefitinib in H1650: a lung cancer cell line with EGFR mutation and primary resistance to EGFR-TK inhibitors. Cancer Chemother Pharmacol 2010;66:381-8. [Crossref] [PubMed]
  75. Gong Y, Yao E, Shen R, et al. High expression levels of total IGF-1R and sensitivity of NSCLC cells in vitro to an anti-IGF-1R antibody (R1507). PLoS One 2009;4:e7273. [Crossref] [PubMed]
  76. Drilon A, Oxnard GR, Tan DSW, et al. Efficacy of Selpercatinib in RET Fusion-Positive Non-Small-Cell Lung Cancer. N Engl J Med 2020;383:813-24. [Crossref] [PubMed]
  77. Lipson D, Capelletti M, Yelensky R, et al. Identification of new ALK and RET gene fusions from colorectal and lung cancer biopsies. Nat Med 2012;18:382-4. [Crossref] [PubMed]
  78. Thein KZ, Velcheti V, Mooers BHM, et al. Precision therapy for RET-altered cancers with RET inhibitors. Trends Cancer 2021;7:1074-88. [Crossref] [PubMed]
  79. Zhu VW, Klempner SJ, Ou SI. Receptor Tyrosine Kinase Fusions as an Actionable Resistance Mechanism to EGFR TKIs in EGFR-Mutant Non-Small-Cell Lung Cancer. Trends Cancer 2019;5:677-92. [Crossref] [PubMed]
  80. Wang C, Zhang Z, Sun Y, 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. [Crossref] [PubMed]
  81. Rotow J, Patel JD, Hanley MP, 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;29:2979-87. [Crossref] [PubMed]
  82. Piotrowska Z, Isozaki H, Lennerz JK, et al. Landscape of Acquired Resistance to Osimertinib in EGFR-Mutant NSCLC and Clinical Validation of Combined EGFR and RET Inhibition with Osimertinib and BLU-667 for Acquired RET Fusion. Cancer Discov 2018;8:1529-39. [Crossref] [PubMed]
  83. Urbanska EM, Sørensen JB, Melchior LC, et al. Durable Response to Combined Osimertinib and Pralsetinib Treatment for Osimertinib Resistance Due to Novel Intergenic ANK3-RET Fusion in EGFR-Mutated Non-Small-Cell Lung Cancer. JCO Precis Oncol 2022;6:e2200040. [Crossref] [PubMed]
  84. Wood DE. National Comprehensive Cancer Network (NCCN) Clinical Practice Guidelines for Lung Cancer Screening. Thorac Surg Clin 2015;25:185-97. [Crossref] [PubMed]
  85. Chang A. Chemotherapy, chemoresistance and the changing treatment landscape for NSCLC. Lung Cancer 2011;71:3-10. [Crossref] [PubMed]
  86. Yang Z, Tam KY. Combination Strategies Using EGFR-TKi in NSCLC Therapy: Learning from the Gap between Pre-Clinical Results and Clinical Outcomes. Int J Biol Sci 2018;14:204-16. [Crossref] [PubMed]
  87. Hosomi Y, Morita S, Sugawara S, et al. Gefitinib Alone Versus Gefitinib Plus Chemotherapy for Non-Small-Cell Lung Cancer With Mutated Epidermal Growth Factor Receptor: NEJ009 Study. J Clin Oncol 2020;38:115-23. [Crossref] [PubMed]
  88. Planchard D, Feng PH, Karaseva N, et al. Osimertinib plus platinum-pemetrexed in newly diagnosed epidermal growth factor receptor mutation-positive advanced/metastatic non-small-cell lung cancer: safety run-in results from the FLAURA2 study. ESMO Open 2021;6:100271. [Crossref] [PubMed]
  89. Watanabe S, Yamaguchi OU, Masumoto AI, et al. Phase I Study Evaluating the Combination of Afatinib with Carboplatin and Pemetrexed After First-line EGFR-TKIs. Anticancer Res 2018;38:4699-704. [Crossref] [PubMed]
  90. Zhao Y, Liu J, Cai X, et al. Efficacy and safety of first line treatments for patients with advanced epidermal growth factor receptor mutated, non-small cell lung cancer: systematic review and network meta-analysis. BMJ 2019;367:l5460. [Crossref] [PubMed]
  91. Wu Q, Luo W, Li W, et al. First-Generation EGFR-TKI Plus Chemotherapy Versus EGFR-TKI Alone as First-Line Treatment in Advanced NSCLC With EGFR Activating Mutation: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Front Oncol 2021;11:598265. [Crossref] [PubMed]
  92. Gijtenbeek RGP, van der Noort V, Aerts JGJV, et al. Randomised controlled trial of first-line tyrosine-kinase inhibitor (TKI) versus intercalated TKI with chemotherapy for EGFR-mutated nonsmall cell lung cancer. ERJ Open Res 2022;8:00239-2022. [Crossref] [PubMed]
  93. Yan H, Li H, Li Q, et al. The Efficacy of Synchronous Combination of Chemotherapy and EGFR TKIs for the First-Line Treatment of NSCLC: A Systematic Analysis. PLoS One 2015;10:e0135829. [Crossref] [PubMed]
  94. Baker S, Dahele M, Lagerwaard FJ, et al. A critical review of recent developments in radiotherapy for non-small cell lung cancer. Radiat Oncol 2016;11:115. [Crossref] [PubMed]
  95. Wang X, Zeng Z, Cai J, et al. Efficacy and acquired resistance for EGFR-TKI plus thoracic SBRT in patients with advanced EGFR-mutant non-small-cell lung cancer: a propensity-matched retrospective study. BMC Cancer 2021;21:482. [Crossref] [PubMed]
  96. Zheng L, Wang Y, Xu Z, et al. Concurrent EGFR-TKI and Thoracic Radiotherapy as First-Line Treatment for Stage IV Non-Small Cell Lung Cancer Harboring EGFR Active Mutations. Oncologist 2019;24:1031-e612. [Crossref] [PubMed]
  97. Akamatsu H, Murakami H, Harada H, et al. Gefitinib With Concurrent Thoracic Radiotherapy in Unresectable Locally Advanced NSCLC With EGFR Mutation; West Japan Oncology Group 6911L. J Thorac Oncol 2021;16:1745-52. [Crossref] [PubMed]
  98. Jia W, Guo H, Jing W, et al. An especially high rate of radiation pneumonitis observed in patients treated with thoracic radiotherapy and simultaneous osimertinib. Radiother Oncol 2020;152:96-100. [Crossref] [PubMed]
  99. D'Antonio C, Passaro A, Gori B, et al. Bone and brain metastasis in lung cancer: recent advances in therapeutic strategies. Ther Adv Med Oncol 2014;6:101-14. [Crossref] [PubMed]
  100. Khan M, Lin J, Liao G, et al. Whole Brain Radiation Therapy Plus Stereotactic Radiosurgery in the Treatment of Brain Metastases Leading to Improved Survival in Patients With Favorable Prognostic Factors. Front Oncol 2019;9:205. [Crossref] [PubMed]
  101. Weber B, Winterdahl M, Memon A, et al. Erlotinib accumulation in brain metastases from non-small cell lung cancer: visualization by positron emission tomography in a patient harboring a mutation in the epidermal growth factor receptor. J Thorac Oncol 2011;6:1287-9. [Crossref] [PubMed]
  102. Togashi Y, Masago K, Masuda S, et al. Cerebrospinal fluid concentration of gefitinib and erlotinib in patients with non-small cell lung cancer. Cancer Chemother Pharmacol 2012;70:399-405. [Crossref] [PubMed]
  103. Zhao J, Chen M, Zhong W, et al. Cerebrospinal fluid concentrations of gefitinib in patients with lung adenocarcinoma. Clin Lung Cancer 2013;14:188-93. [Crossref] [PubMed]
  104. Fan Y, Huang Z, Fang L, et al. A phase II study of icotinib and whole-brain radiotherapy in Chinese patients with brain metastases from non-small cell lung cancer. Cancer Chemother Pharmacol 2015;76:517-23. [Crossref] [PubMed]
  105. Zhou K, Cai X, Wang X, et al. Efficacy and safety of WBRT+EGFR-TKI versus WBRT only in the treatment of NSCLC patients with brain metastasis: An updated meta-analysis. Thorac Cancer 2022;13:563-70. [Crossref] [PubMed]
  106. He ZY, Li MF, Lin JH, et al. Comparing the efficacy of concurrent EGFR-TKI and whole-brain radiotherapy vs EGFR-TKI alone as a first-line therapy for advanced EGFR-mutated non-small-cell lung cancer with brain metastases: a retrospective cohort study. Cancer Manag Res 2019;11:2129-38. [Crossref] [PubMed]
  107. Marampon F, Gelibter AJ, Cicco PR, et al. Safety and efficacy of combining afatinib and whole-brain radiation therapy in treating brain metastases from EGFR-mutated NSCLC: a case report and literature review. BJR Case Rep 2022;8:20200134. [Crossref] [PubMed]
  108. Riley RS, June CH, Langer R, et al. Delivery technologies for cancer immunotherapy. Nat Rev Drug Discov 2019;18:175-96. [Crossref] [PubMed]
  109. Osmani L, Askin F, Gabrielson E, et al. Current WHO guidelines and the critical role of immunohistochemical markers in the subclassification of non-small cell lung carcinoma (NSCLC): Moving from targeted therapy to immunotherapy. Semin Cancer Biol 2018;52:103-9. [Crossref] [PubMed]
  110. Mino-Kenudson M. Programmed cell death ligand-1 (PD-L1) expression by immunohistochemistry: could it be predictive and/or prognostic in non-small cell lung cancer? Cancer Biol Med 2016;13:157-70. [Crossref] [PubMed]
  111. Ghosh C, Luong G, Sun Y. A snapshot of the PD-1/PD-L1 pathway. J Cancer 2021;12:2735-46. [Crossref] [PubMed]
  112. Yu W, Hua Y, Qiu H, et al. PD-L1 promotes tumor growth and progression by activating WIP and β-catenin signaling pathways and predicts poor prognosis in lung cancer. Cell Death Dis 2020;11:506. [Crossref] [PubMed]
  113. Gibbons DL, Chow LQ, Kim DW, et al. 57O Efficacy, safety and tolerability of MEDI4736 (durvalumab [D]), a human IgG1 anti-programmed cell death-ligand-1 (PD-L1) antibody, combined with gefitinib (G): a phase I expansion in TKI-naïve patients (pts) with EGFR mutant NSCLC. J Thorac Oncol 2016;11:abstr S79.
  114. Oxnard GR, Yang JC, Yu H, et al. TATTON: a multi-arm, phase Ib trial of osimertinib combined with selumetinib, savolitinib, or durvalumab in EGFR-mutant lung cancer. Ann Oncol 2020;31:507-16. [Crossref] [PubMed]
  115. Salvi S, Fontana V, Boccardo S, et al. Evaluation of CTLA-4 expression and relevance as a novel prognostic factor in patients with non-small cell lung cancer. Cancer Immunol Immunother 2012;61:1463-72. [Crossref] [PubMed]
  116. Chalmers AW, Patel S, Boucher K, et al. Phase I Trial of Targeted EGFR or ALK Therapy with Ipilimumab in Metastatic NSCLC with Long-Term Follow-Up. Target Oncol 2019;14:417-21. [Crossref] [PubMed]
  117. Puri S, Patel S, Chalmers A, et al. P15. 02 Phase Ib Study to Evaluate the Safety and Efficacy of Osimertinib in Combination with Ipilimumab in EGFR Mutated NSCLC. J Thorac Oncol 2021;16:abstr S344.
  118. Peravali M, Wang H, Kim C, et al. Combined Inhibition of EGFR and VEGF Pathways in Patients with EGFR-Mutated Non-Small Cell Lung Cancer: A Systematic Review and Meta-Analysis. Curr Oncol Rep 2020;22:119. [Crossref] [PubMed]
  119. Zhou Q, Xu CR, Cheng Y, et al. Bevacizumab plus erlotinib in Chinese patients with untreated, EGFR-mutated, advanced NSCLC (ARTEMIS-CTONG1509): A multicenter phase 3 study. Cancer Cell 2021;39:1279-1291.e3. [Crossref] [PubMed]
  120. Kuo CS, Chiu TH, Tung PH, et al. Afatinib Treatment Alone or with Bevacizumab in a Real-World Cohort of Non-Small Cell Lung Cancer Patients with Epidermal Growth Factor Receptor Mutation. Cancers (Basel) 2022;14:316. [Crossref] [PubMed]
  121. Soo RA, Han JY, Dafni U, et al. A randomised phase II study of osimertinib and bevacizumab versus osimertinib alone as second-line targeted treatment in advanced NSCLC with confirmed EGFR and acquired T790M mutations: the European Thoracic Oncology Platform (ETOP 10-16) BOOSTER trial. Ann Oncol 2022;33:181-92. [Crossref] [PubMed]
  122. Kenmotsu H, Wakuda K, Mori K, et al. Randomized Phase 2 Study of Osimertinib Plus Bevacizumab Versus Osimertinib for Untreated Patients With Nonsquamous NSCLC Harboring EGFR Mutations: WJOG9717L Study. J Thorac Oncol 2022;17:1098-108. [Crossref] [PubMed]
Cite this article as: Zhang Q, Wang R, Xu L. Clinical advances in EGFR-TKI combination therapy for EGFR-mutated NSCLC: a narrative review. Transl Cancer Res 2023;12(12):3764-3778. doi: 10.21037/tcr-23-956

Download Citation