Treatment of advanced non-small cell lung cancer with driver mutations: current applications and future directions

Jia Zhong , Hua Bai , Zhijie Wang , Jianchun Duan , Wei Zhuang , Di Wang , Rui Wan , Jiachen Xu , Kailun Fei , Zixiao Ma , Xue Zhang , Jie Wang

Front. Med. ›› 2023, Vol. 17 ›› Issue (1) : 18 -42.

PDF (1728KB)
Front. Med. ›› 2023, Vol. 17 ›› Issue (1) : 18 -42. DOI: 10.1007/s11684-022-0976-4
REVIEW
REVIEW

Treatment of advanced non-small cell lung cancer with driver mutations: current applications and future directions

Author information +
History +
PDF (1728KB)

Abstract

With the improved understanding of driver mutations in non-small cell lung cancer (NSCLC), expanding the targeted therapeutic options improved the survival and safety. However, responses to these agents are commonly temporary and incomplete. Moreover, even patients with the same oncogenic driver gene can respond diversely to the same agent. Furthermore, the therapeutic role of immune-checkpoint inhibitors (ICIs) in oncogene-driven NSCLC remains unclear. Therefore, this review aimed to classify the management of NSCLC with driver mutations based on the gene subtype, concomitant mutation, and dynamic alternation. Then, we provide an overview of the resistant mechanism of target therapy occurring in targeted alternations (“target-dependent resistance”) and in the parallel and downstream pathways (“target-independent resistance”). Thirdly, we discuss the effectiveness of ICIs for NSCLC with driver mutations and the combined therapeutic approaches that might reverse the immunosuppressive tumor immune microenvironment. Finally, we listed the emerging treatment strategies for the new oncogenic alternations, and proposed the perspective of NSCLC with driver mutations. This review will guide clinicians to design tailored treatments for NSCLC with driver mutations.

Keywords

non-small cell lung cancer / driver mutations / treatment strategy / resistant mechanism / immune-checkpoint inhibitors

Cite this article

Download citation ▾
Jia Zhong, Hua Bai, Zhijie Wang, Jianchun Duan, Wei Zhuang, Di Wang, Rui Wan, Jiachen Xu, Kailun Fei, Zixiao Ma, Xue Zhang, Jie Wang. Treatment of advanced non-small cell lung cancer with driver mutations: current applications and future directions. Front. Med., 2023, 17(1): 18-42 DOI:10.1007/s11684-022-0976-4

登录浏览全文

4963

注册一个新账户 忘记密码

1 Introduction

Lung cancer is the leading cause of cancer-related death worldwide. Improved understanding of driver mutations of non-small cell lung cancer (NSCLC) has led to more biomarker-directed treatment for patients with advanced stages. The expanding number of drugs targeting these driver mutations offers more opportunity to improve patient’s survival benefit.

To date, NSCLCs, especially those with non-squamous histology, are recommended for testing epidermal growth factor receptor (EGFR) mutations, anaplastic lymphoma kinase (ALK) gene rearrangements, ROS proto-oncogene receptor tyrosine kinase 1 (ROS-1) rearrangements, B-raf proto-oncogene (BRAF) mutations, rearranged during transfection (RET) fusions, Met proto-oncogene (MET) amplification and exon 14 skipping alterations, neurotrophic receptor tyrosine kinase (NTRK) gene fusions, and immunohistochemistry (IHC) testing for the programmed death receptor-ligand 1 (PD-L1) expression.

EGFR-activating mutations are the most common driver mutations in NSCLC. Targeted therapies included first-generation epidermal growth factor inhibitor tyrosine kinase inhibitors (EGFR-TKIs), erlotinib, gefitinib, and icotinib; second-generation pan-human epidermal growth receptor (HER) family inhibitor, afatinib and dacotinib; and third-generation EGFR-TKI, osimertinib, that inhibits both EGFR-sensitive mutations and resistant mutation EGFR T790M [13]. ALK inhibitors included first-generation, crizotinib; second generation, ceritinib, alectinib, and brigatinib; and third-generation, lorlatinib, with increasing capacity for ALK inhibition generation-by-generation [4,5]. The resistance caused by secondary ALK mutations can be overcome by next-generation ALK-TKIs [6]. Moreover, alectinib, brigatinib, and lorlatinib are all recommended as first-line treatment choice for ALK-rearranged NSCLC for their superior survival compared with crizontinb [79]. Crizotinib, ceritinib, brigatinib, and lorlatinib can also inhibit ROS-1 [10,11]. BRAF inhibitor dabrafenib combined with MEK inhibitor trametinib is now the recommended treatment for BRAF V600E-mutated NSCLC attributing to improved outcomes compared with vemurafenib or dabrafenib monotherapy [12]. Drugs targeting MET aberrations with different binding modes included type Ia, crizotinib; type Ib, tepotinib, capmatinib, and savolitinib; and type II, cabozantinib. Entrectinib and larotrectinib are the current standard treatment for NTRK fusion-positive NSCLC [13]. Kirsten ratsarcoma viral oncogene homolog (KRAS) mutations occur in ~20%–30% of patients with NSCLC. Recently, several small molecules including sotorasib (AMG510), adagrasib (MRTX-849) have been developed to specifically target KRAS G12C [14].

Although drugs targeting oncogenic driver mutations have significantly improved survival but their long-term responses are still uncommon for most patients, the emergence of acquired drug resistance is inevitable. Therefore, exploration and understanding of the resistance mechanism of target therapy are important to enhance the clinical outcomes and ultimately increase the treatment rate of NSCLC. Meanwhile, improved understanding of biological characteristics in various molecular subtypes of each driver mutation helps explore new classified treatment strategies. Until recently, multiomics analysis has ushered in a new era of precision targeted therapy in lung cancer and led to a deeper understanding of the underlying resistant mechanism. All these scientific and clinical progresses ultimately lead to improved survival in NSCLC and achieve more refined individualized treatment.

We searched PubMed for articles describing clinical trials and reviews of medical therapies for NSCLC with driver mutations form January 1, 2013 to April 1, 2022. The guidelines from major professional societies were also reviewed.

2 Classified management of NSCLC with driver mutation

2.1 Stratification by gene subtypes

EGFR mutations are the most common driver mutations in NSCLC, contributing to approximately 50% of patients with NSCLC in Asians and 15% in Caucasians [15]. Exon 19 deletion (19del) and L858R mutation in exon 21 (L858R) are the major subtypes of EGFR-sensitive mutations. EGFR exon 19 deletion was associated with better response to EGFR-TKI treatment than L858R mutation [16]. A meta-analysis including studies comparing EGFR-TKIs with chemotherapy as the first-line treatment revealed that the benefit of EGFR-TKI versus chemotherapy was 50% higher for patients with exon 19 deletion (hazard ratio (HR) 0.24, 95% confidence interval (CI) 0.20–0.29) than those with exon 21 L858R alternation (HR 0.48, 95% CI 0.39–0.58, P interaction < 0.001) [17]. In the pooled analysis of the LUX-Lung-3 and 6 studies, two phase 3 trials comparing afatinib and chemotherapy, the overall survival (OS) improvement was significant in patients with exon 19 deletion but not in those with exon 21 L858R mutation [18]. In the FLAURA study, the median OS was significantly prolonged in the first-line osimertinib group than the first-generation EGFR-TKI comparator. However, the magnitude of OS benefit varied among the two EGFR mutation subtypes, with the HRs close to 1.00 (95% CI 0.71–1.40) in the L858R subgroup and 0.68 (95% CI 0.51–0.90) in the 19-deletion subgroup [19]. JO22567 and NEJ026 showed that the bevacizumab plus erlotinib significantly improved progression-free survival (PFS) than erlotinib alone in patients with EGFR-positive NSCLC. Subgroup analyses by EGFR mutation subtype suggested that median PFS was longer in patients with L858R mutations in the combination therapy group than those in the erlotinib group (HR 0.57, 95% CI 0.33–0.97). No differences were found between treatment groups in patients with EGFR exon 19 deletions (HR 0.69, 95% CI 0.41–1.16) [20]. Similar results were verified in CTONG 1509, a similar design study in the Chinese population [21]. The INCRESE study showed that double-dose icotinib improved the median PFS (12.9 vs. 9.2 months, HR 0.75, 95% CI 0.53–1.05) and overall response rate (ORR) (73% vs. 48%) in patients with NSCLC harboring 21-L858R mutation compared with the routine dose with acceptable tolerability [22]. All of these findings suggest fundamental differences in biological characteristics and treatment responsiveness between these two subtypes of activating EGFR mutations. Therefore, for patients with EGFR exon 19 deletion, second- or third-generation EGFR-TKI monotherapy is sufficient; however, for patients with EGFR L858R mutation, more aggressive treatments including EGFR-TKIs combined with anti-angiogenesis therapy or dose increase might better extend the PFS and OS.

Most prospective clinical trials of EGFR-TKIs enrolled NSCLC patients with common (19del or L858R) mutations. However, 10%–20% of EGFR mutant NSCLC harbor uncommon mutations, which are less sensitive to EGFR-TKIs treatment. Second- and third-generation EGFR-TKIs have a broader activity profile across uncommon EGFR mutations [23]. A combined analysis of LUX-Lung 2/3/6 indicated that afatinib was active in NSCLC with uncommon EGFR mutations, especially G719X, L861Q, and S768I but less active in de novo T790M and exon 20 insertions [24]. A recent clinical trial indicated that patients harboring uncommon EGFR mutations including exon 20 insertion were also sensitive to osimertinib. Early clinical activity in EGFR/HER2 exon 20 has been observed with the dual EGFR/HER2-TKIs, poziotinib, and TAK-788 [25]. Amivantamab, a monoclonal bispecific anti-EGFR-MET antibody, showed an ORR of 40% and PFS of 8.3months for patients with EGFR exon 20 insertion mutant NSCLC with prior platinum-based chemotherapy in the phase I clinical trial, which was certified by the United States Food and Drug Administration as breakthrough therapy for EGFR exon 20 insertion mutant NSCLC [26,27]. A recent study also found that NSCLC with uncommon EGFR mutations associated with a significantly lower incidence of acquired T790M mutation and benefited significantly less from subsequent osimertinib treatment than common EGFR mutations [28].

The mutation abundance also affects the therapeutic efficacy of EGFR-TKI. Studies indicated both tissue and plasma abundance of EGFR driver mutation affected clinical response and PFS of EGFR-TKIs [29,30]. Meanwhile, the mutation abundance of EGFR T790M could also predict the efficacy of osimertinib [31].

Besides EGFR, other gene alternations also need to stratify patients according to gene subtypes, such as ALK rearrangement. Currently, eight EML4-ALK variants (V) have been identified with a number (1, 2, 3a/b, 4', 5a/b, 5′, 7, 8), among them EML4-ALK variants 1 and 3 being the two predominant variants. Different clinical response to ALK-TKIs between the “short” (V3 and V5) and “long” (V1, V2, V5′, V7, and V8) EML4-ALK variants has been reported. According to clinical data concerning crizotinib or lorlatinib, patients with V3a/b seem to have had significantly shorter PFS than did patients with V1. However, these results conflict with those of other studies with crizotinib or alectinib that report insignificant differences in PFS among patients with V1, V2, and V3 [32]. Meanwhile, V3 was associated with worse overall survival in ALK-positive NSCLC [33]. Additionally, ALK G1202R is more prone to develop among EML4-ALK V3 after use of ALK TKIs [34].

Generally, BRAF alterations can be categorized into three classes. Class I BRAF mutations including BRAF V600 mutations function as RAS-independent monomers, which represent most of BRAF alterations observed in cancer; non-V600 BRAF mutants can be divided into two categories, class II mutations, including K601E, L597Q, and G469A, signal as RAS-independent activated dimers, and class III mutations, including D594 and G466 mutants, which are kinase impaired but increase signaling through enhanced RAS binding and signal as dimers [35,36]. Class I BRAF mutation well response to BRAF and MEK dual inhibition. However, the treatment of NSCLC harboring non-V600 mutations is challenging because of functional heterogeneity and lack of clinical data, though few study indicated that certain non-V600 BRAF alternations (such as L597R mutation, class II mutation) also have durable response to MEK and BRAF inhibitors [37]. These distinct responses of different classes of BRAF mutations have important implications for future classified management of BRAF mutant NSCLC.

RET rearrangements are an emerging targetable driver mutation in NSCLC. The most common fusion partner was the kinesin family 5B gene (KIF5B), followed by coiled-coil domain containing 6 (CCDC6). Due to the low prevalence of RET fusions, there is only limited data about the clinical characteristics and outcomes of RET-rearranged NSCLC. Patients with CCDC6-RET fusion may have a better prognosis [38] and longer PFS for chemotherapy [39] compared with KIF5B-RET and other subtypes. For target therapy, ORR was similar with KIF5B and CCDC6, but lower in other RET fusions in ARROW study [40].

Alterations in MET include MET protein overexpression, MET gene amplification, and MET exon 14 skipping mutations or activating mutations. MET overexpression is related to poor clinical outcome in NSCLCs but its role as a therapeutic marker remains controversial. MET amplification has been regarded as a therapeutic biomarker, with clinical data demonstrating that the high MET gene copy number indicated a high response rate to targeted therapies. MET exon 14 skipping mutations also indicate poor outcomes in NSCLC and are presently the most promising driver mutation for the use of MET-TKIs. As we mentioned before, there are three subtypes of MET-TKIs and response differently to the MET mutations: type I MET inhibitors bind to the activated ATP-pocket of MET, and are subdivided into Ia and Ib; type II inhibitors bind the ATP-pocket in the inactive state; type III inhibitors bind to allosteric sites distinct from the ATP binding site. Mutations in the MET activation loop D1228 and Y1230 confer resistance to type Ia MET-TKIs while sensitive to type II kinase inhibitors. The solvent front mutation G1163R mediates resistance only to type Ia inhibitor crizotinib but not to type Ib or type II MET inhibitors. Conversely, mutations in residues L1195 and F1200 confer resistance to type II MET inhibitors. These findings provide rationality for sequential or combination therapy to avoid resistance to MET TKIs [4143].

2.2 Stratification by concomitant mutation

EGFR is a member of the ErbB family of receptor tyrosine kinases. Its downstream pathways include PI3K-AKT-mTOR, JAK-STAT, RAF-MEK-ERK, and other pathways. Co-mutations in downstream and other ErbB family members can spur the cell growth through signal transduction, eventually resulting in tumor proliferation and even metastasis [44]. Clinical data also indicated the concomitant genetic alterations associated with poorer response to EGFR-TKI treatment among the EGFR mutant NSCLC [45]. Some co-mutations such as TGFBR1, MTOR, and RNF43 might indicate primary resistance to EGFR-TKIs [46]. In the BENEFIT study [47], a prospective study using circulating tumor DNA (ctDNA) as direct first-line EGFR-TKI treatment, patients with tumor suppressor co-mutations and tumors with other driver mutations had shorter PFS (9.3 (7.6–10.5) vs. 4.0 (2.4–9.3) months vs. 13.2 (11.3–15.2)) and OS (21.7 (19.3–27.0)) vs. 15.5 ((10.5–33.7) months vs. 32.0 (29.2–41.5)) than those with pure EGFR-sensitive mutation [48]. Preclinical data suggest that EGFR-TKIs combined with chemotherapy may act synergistically to restrict the development of acquired resistance [49]. Meanwhile, clinical data also indicated that the adverse events are manageable [50]. For patients with concomitant mutation, the treatment mode like that of the NEJ009 study, EGFR-TKIs combined with chemotherapy [51], might improve the prognosis of patient with concomitant mutations. Some clinical trials aiming to assess the efficacy of combined EGFR-TKIs and chemotherapy in NSCLC with co-mutations are ongoing (ChiCTR2000032354).

Occasionally, tumors have T790M coexistence with EGFR-activating mutations at the time of diagnosis. Using more sensitive methods such as DNA mass array, the occurrence of T790M in TKI-naïve patients was higher than that in direct sequencing (25.2% vs. 2.8%, P < 0.001) [52]. Naturally, third-generation EGFR-TKIs are the recommended choice of patients with de novo T790M.

Besides EGFR concomitant mutations, EGFR amplification was also regarded as an EGFR-dependent resistant mechanism to EGFR-TKIs [53]. EGFR amplification is more likely to arise in cases with resistance to third generation of EGFR-TKIs [54], and accounts for approximately 20% of the resistant mechanism. The EGFR-MET bispecific antibody amivantamab in combination of third-generation EGFR-TKIs might improve the outcome of patient with EGFR amplification and EGFR sensitive mutations. The clinical trial that is called the MARIPOSA study (NCT04487080) to compare the antitumor activity of amivantamab + lazertinib versus osimertinib alone in patients with EGFR mutant NSCLC first line treatment is ongoing [55].

The concomitant sensitive mutations rarely occurred. The frequency of EGFR/ALK or EGFR/ROS-1 co-alternations was about 1% [56]. Based on the published data and case report, EGFR/ALK co-altered NSCLC still showed response to EGFR-TKIs [57,58], though the EGFR-sensitizing mutant NSCLC with other driver mutations exhibited shorter PFS (4.0 versus 13.2 months) and OS (15.5 versus 32.0 months) compared with those with EGFR-sensitive mutation alone [48,59]. The co-mutation of EGFR and MET (2% prevalence) or ERBB2 (4% prevalence) amplification was also associated with significantly shorter progression-free survival with first-line 1st or 2nd generation EGFR-TKIs therapy [60]. Generally, the first-line EGFR-TKIs might be a reasonable care for the NSCLC with EGFR and other driver mutations. The frequency of the co-alterations, the sensitivity of the detection methods, and the phosphorylation levels of the driver gene and its downstream proteins should be considered when making treatment choice [61].

Like EGFR mutations, concomitant mutations in the non-EGFR mutation-driven NSCLC also mostly indicated poor duration of response. For ALK fusion NSCLC, the presence of TP53 mutations is a poor predictive and prognostic factor. Though lacks of clinical data, pre-clinical study provided insight that the combination of ALK-TKIs and proteasome potentially leads to TP53-independent apoptosis [34]. The co-occurrent PIK3CA E542K and EML4-ALK V3 was also reported to be resistant to ALK-TKIs [62]. For RET-rearranged NSCLC, patient with concomitant TP53 mutation showed shorter overall survival than those without [63]. Specially, patient with STK11 mutation, KRAS G12C mutation and wild type KEAP1 benefit more from the sotorasib therapy [64].

2.3 Stratification by dynamic alternation

The dynamic gene mutation changes are also key predictive factors for target therapy. Liquid biopsy identifying molecular alterations in plasma ctDNA makes the dynamic monitoring possible. In the BENEFIT study, 147 (88%) patients exhibited EGFR mutation clearance in ctDNA at week 8, and the median PFS was longer for these patients than those with EGFR mutations that persisted at week 8 (11.0 months (95% CI 9.43–12.85) vs. 2.1 months (1.81–3.65); HR 0.14, 95% CI 0.08–0.23; P < 0.0001) [47]. Patients with persistent activating EGFR mutations in plasma ctDNA after osimertinib initiation also indicated worse prognosis, either in the first-line setting [65] or T790M-positive patients as the second-line treatment [66]. Patients without detectable EGFR mutation in the pretreatment ctDNA possessed the best prognosis from the sequential TKI treatment [67].

Patients with ctDNA nonclearance at the first visit might require combined therapy because of the limited survival benefit of EGFR-TKI monotherapy. A clinical trial that investigates the efficacy and safety of osimertinib plus carboplatin/pemetrexed versus osimertinib monotherapy in metastatic EGFR mutant NSCLC patients with EGFR mutation persistence in ctDNA 3 weeks after the first-line osimertinib is ongoing (NCT04769388).

Therefore, for EGFR, the target therapy should be stratified based on diverse mutation subtypes, concomitant mutations, and dynamic ctDNA changes. With the in-depth understanding of other driver mutations, this stratified treatment mode can be an effective reference to other target therapy.

2.4 Stratification by special metastasis site

Brain or leptomeningeal metastasis is a generally negative prognostic factor and is refractory to treatment due to the blood–brain barrier. Second- or third-generation EGFR-TKIs [52] or ALK-TKIs are better options for those patients. Second- or third-generation EGFR and ALK inhibitors had higher intracranial ORR and disease control rate (DCR) in patients refractory to first-generation TKIs; thus, the next-generation EGFR/ALK-TKIs are currently replacing first-generation TKIs as the first-line treatment, especially for NSCLC with central nerve system (CNS) metastasis [68]. Stereotactic radiosurgery and whole-brain radiotherapy are also the treatment options for patients with CNS metastasis.

Oligo-metastatic disease represents a special subtype of metastatic NSCLC that required local treatment [69]. A series of phase II clinical trials confirmed that local consolidative therapy for patients with oligo-metastatic NSCLC did not progress after initial systemic therapy, which more significantly improved PFS than those without local therapy [70,71]. Moreover, for oligo-progressive conditions during target therapy, local ablation of these resistant lesions using either surgery or radiation can prolong the long-term response of the existing target therapy, with an average time to next progression of 6–7 months [72,73].

2.5 Stratification by multinomic profiles

With the advent of new genomics, transcriptomics, metabolomics, and proteomics technology, integrated genomic studies have fundamentally contributed to the definition of multinomics landscape of NSCLC. Copy number alterations represent one of the key genetic alterations and included two types: large-scale copy number gain or loss and focal events of gene amplification or loss [74]. The large-scale copy number gain or loss is consistent in most lung adenocarcinoma [75], whereas some focal events were associated with negative outcomes, such as KRAS gene amplification and MET focal gains [74]. A previous study also indicated the use of DNA-sequencing, RNA-sequencing, and immunological biomarkers for patients matched to targeted therapies and to identify new potential cancer driver genes [76,77]. Mutational signature may also help identify patients. The APOBEC signature was associated with early-onset EGFR wild-type lung cancer in women, which is in line with the finding that the APOBEC signature was associated with better immunotherapeutic response [78].

3 Resistance mechanisms and coping strategies

Although the target therapy improves survival in NSCLC with driver mutations, only a part of patients respond to these agents and the benefit is temporary. Recognition of the resistance complexity to targeted therapeutics is necessary to understand the tumor biological behavior and to design optional therapeutic strategies.

Based on the duration of disease control, resistance to targeted agents can be classified into intrinsic and acquired resistance [79]. Primary resistance is defined as resistance in NSCLC with sensitive mutations that progressed within 90 days (3 months) and without showing any evidence of objective responses while receiving target therapy [80]. The intrinsic resistance might be associated with insensitive driver mutations. For EGFR-TKIs, intrinsic-resistant mechanisms include EGFR uncommon mutations, co-mutations that activate downstream or bypass signaling pathways, the presence of germline Bcl-2-interacting mediator of cell death (BIM, also known as BCL2L11), or activation of NF-κB (nuclear factor-κB) that impairs the apoptosis process in EGFR-TKI treatment. Acquired resistance refers to tumors that initially responded to the target therapy (usually > 6 months) but ultimately progressed based on Response Evaluation Criteria in Solid Tumors [81]. Acquired resistance increases both from the selection for pre-existing genetic alterations within a heterogeneous tumor and from the acquisition of new mutations under the therapeutic selective pressure [79].

Based on the disease failure mode, disease progression in the target therapy can be classified into three subgroups: dramatic, gradual, and local progression. Patients with NSCLC with gradual progression have longer PFS and OS compared with those with local and dramatic progression, and the TKI continuation is superior to switching to chemotherapy in a subsequent setting for patients with gradual progression. For patients with local progressive disease, continued use of the target therapy plus local therapy is recommended. Conversely, studies demonstrated a better survival with chemotherapy instead of continued TKIs for patients with dramatic progression [82].

Depending on the resistant mechanism, the two major pathways of therapeutic failure are additional genomic mutations in the targeted oncogene (target-dependent resistance) and alternations leading to bypass or downstream activation (target-independent resistance) [83].

3.1 Target-dependent resistance

The target-dependent resistance mutations include second-site mutations, oncogene amplification or loss, limiting the ability of a drug to inhibit the target activity. The second-site mutations include T790M mutation that leads to acquired resistance to first- and second-generation EGFR-TKIs and C797S mutation that mediates the resistance to third-generation EGFR-TKIs. A recent study separated EGFR mutations into four distinct subgroups based on sensitivity and structural changes, indicating that the on-target mutation might cause EGFR structural changes, thus causing different outcomes following treatment with EGFR inhibitors [84]. The on-target mutation is the most common resistant mechanism for third-generation EGFR-TKIs, such as C797S, G796S, and L792F/H mutations that abolish the covalent binding or confer structural steric hindrance of osimertinib to the EGFR protein. “Fourth-generation” EGFR-TKIs such as EAI045, JBJ-04-125-02, and BLU-945 are being developed to inhibit these triple mutations (i.e., del19 or L858R/T790M/C797S), yet remain at the early clinical stages of development [85]. The EGFR/MET bispecific antibody amivantamab has demonstrated activity against a myriad of compound EGFR mutations [86].

Meanwhile, ALK L1196M [87] and ROS1 L2026M [88] that sterically hinder TKI binding also lead to resistance to first-generation ALK-TKIs. Similarly, RET G810X mutation is responsible for resistance to RET-selective inhibitors, such as selpercatinib (LOXO-292) and pralsetinib (BLU-667) [89]. Both KRAS R68S and Y96C/D/S drug binding resistance mutations show high-level resistance to both sotorasib and adagrasib [9092]. EGFR amplification and loss of EGFR T790M mutation have been reported to be resistant to third-generation EGFR-TKIs [93]. Similarly, ALK amplification and copy number gain mediate resistance to crizotinib [94]. KRAS G12C amplification has been reported at resistance to adagrasib (Tab.1) [91].

Moreover, for NSCLC with EML4-ALK fusion, the eight EML4-ALK variants also demonstrate a differential clinical response to ALK-TKIs, with variants 3 and 5, indicating poor responses and short PFS [95].

3.2 Target-independent resistance

Target-independent resistance occurs through the parallel activation or downstream signaling pathways using the driver oncoprotein [96]. Taking EGFR (also known as ERBB1) for example, the alternation in other ERBB family members such as HER2, HER3, and HER4 all results in the activation of three downstream pathways including PI3K-AKT, JAK-STAT, and MAPK pathways [97].

In EGFR-mutated tumors, the MAPK pathway reactivation can occur by acquiring a BRAF mutation (V600E), loss of NF1 gene, and activation of NRAS mutations [98,99]. In preclinical studies, this resistance to third-generation EGFR-TKIs was reversed by combination treatment with MEK and EGFR inhibitors [98]. The PTEN loss and PIK3CA mutations are the most common resistant mechanisms in the PIK3CA-AKT pathway. Inhibitors targeting the PI3K-AKT-mTOR pathway are now in clinical trials and have shown potent efficacy. Autocrine interleukin-6 (IL-6) signaling by tumor cells increases JAK-STAT3 activity, which can be activated as an adaptive response to the EGFR-TKI treatment.

SRC is an oncoprotein implicated in tumor survival and differentiation. SRC activation was reported in EGFR-TKI-resistant NSCLC cell lines, and the addition of SRC inhibitor dasatinib to erlotinib showed early signs of clinical efficacy in patients with EGFR mutant NSCLC [100]. MET and its ligand, hepatocyte growth factor (HGF), can activate MAPK and PI3K-AKT-mTOR signaling. MET amplification and exon 14 skip mutation account for 10%–20% of EGFR-TKI-resistant NSCLC. A series of clinical trials concerning combined EGFR and MET inhibitors are ongoing. Meanwhile, MET variations are also responsible for ALK-TKI resistance in ALK-rearranged NSCLC.

The receptor tyrosine kinase AXL also activates MAPK and PI3K-AKT signaling. The overexpression of AXL in NSCLC resulted in resistance to both EGFR-TKIs and ALK-TKIs [101]. The AXL-TKI is also at the early phase clinical trials. HER2 gene amplification/overexpression and HER2 exon 20 insertion mutations can also lead to bypass activation. Poziotinib and pyrotinib have shown potent inhibitory abilities against HER2 mutant NSCLC; however, patients with HER2 overexpression have shown limited responses to HER2 inhibitors [96,102].

The “Target-independent resistance” mechanism can be subdivided into tumor suppressor gene, other driver gene, cell cycle related gene and others. Acquired bypass resistance mechanisms of KRAS G12C inhibitors included MET and HER2 amplification; activating mutations in NRAS, BRAF, MAP2K1, and RET; oncogenic fusions involving ALK, RET, BRAF, RAF1, and FGFR3; and loss-of-function mutations in NF1 and PTEN [91,103105]. The majority of resistance to selective RET inhibition may be driven by RET-independent resistance, such as acquired MET or KRAS amplification (Tab.1) [106].

3.3 Other resistant mechanisms

3.3.1 Phenotype transformation

Phenotype transformation is a less common mechanism of EGFR-TKI resistance and other target therapies, encompassing the histological transformation of adenocarcinoma to small cell lung cancer (SCLC) and to squamous cell carcinoma (SCC) [83]. The transformed derivatives manifest a more aggressive subtype that can escape from anticancer-targeted therapies. Epithelial-to-mesenchymal transition (EMT) is another phenotypic transformation as a resistance change to both EGFR and ALK-TKI therapies [107], indicating a more invasive phenotype. RB1 and TP53 deficiencies indicated a higher incidence of SCLC transformation [108], and the classical chemotherapy for SCLC is also sensitive to the transformed SCLC [109].

EMT is a cause of both intrinsic and acquired resistance of KRAS G12C inhibitors by activating the PI3K pathway [110]. Moreover, histological transformation from adenocarcinoma to squamous cell carcinoma was observed in patients resistant to adagrasib without determining any genomic resistance mechanisms [90].

3.3.2 Tumor microenvironment (TME)

TME refers to the tumor-surrounded stromal components, including fibroblasts, immune cells, blood vessels, and cytokines, influencing the response to targeted therapy. Coculture of cancer-associated fibroblasts (CAFs) can induce both EMT and resistance to EGFR-TKIs in NSCLC cells [111]. Cytokines, such as HGF, activate the ERK signaling and consequently result in EGFR-TKI resistance [112]. The HGF secretion by CAFs can also lead to ALK-TKI resistance by inducing MET-dependent signaling activation [113]. Furthermore, the VEGF pathway is a key mediator of tumor angiogenesis. In vitro studies have found that VEGF and EGFR pathways share common oncogenic downstream signaling and elevated VEGF contributing to the emergence of EGFR-TKI resistance. In clinical trials, the addition of anti-VEGF therapy to EGFR-TKIs significantly improved the clinical outcomes [114]. Angiogenic factors such as the Tie2 receptor also regulated EGFR downstream signaling PI3K/AKT and/or MAPK pathways [115].

3.3.3 Impaired apoptosis

Germline BIM (also known as BCL2L11) deletion polymorphism impairs the apoptotic response to EGFR-TKI therapy, thus leading to drug resistance [116]. Activation of NF-κB weakens the apoptosis and rescues EGFR mutant NSCLC from EGFR-TKI treatment. The inactivation of NF-κB pathway enhances cell death induced by EGFR-TKIs [117]. A recent study has found that YES-associated protein (YAP) activation mediates resistance to combined EGFR/MEK inhibition. The combination of drugs targeting the YAP pathway, such as transcriptional enhancer associate domain (TEAD) inhibitor, promotes apoptosis of resistant cancer cells [52].

3.3.4 Barriers and drug–drug interactions (DDIs)

Further challenges beyond the former factors include overcoming barriers, such as blood–brain barriers that limit effective drug delivery to CNS metastases. Increasing drug exposure by dose escalation or exploring new-generation TKIs is a possible solution to overcome body barriers. DDIs should also be under consideration. The combination of drugs interacting with cytochrome P450 and P-glycoprotein should be avoided. Proton pump inhibitors also impair the pH-dependent solution, thus reducing drug absorption [118].

3.3.5 Resistant mechanism based on multiomics analysis

By analyzing the adaptive-resistant tumor cells using transcriptomic and metabolomics profiling, drug resistance was found to be correlated with cells in proliferative-metabolic quiescence and susceptible to glutamine deprivation and TGFβ2 inhibition [119]. This study and others [120] support the co-targeting of bioenergetics and mitochondrial priming to suppress the onset of early drug resistant to EGFR-TKIs.

4 ICIs in NSCLC with driver mutations

4.1 Immune-checkpoint inhibitors show limited effectiveness in NSCLC with driver mutations

Patients with EGFR mutation poorly respond to PD-1/PD-L1 inhibitors. In the PACIFIC trial of patients with unresectable, stage III NSCLC, the consolidative durvalumab failed to significantly improve the PFS and OS compared with the placebo group in 6.0% patients with EGFR mutations [164166]. A meta-analysis, including CheckMate 017/057, KEYNOTE-010, POPLAR, and OAK found that nivolumab, pembrolizumab, and atezolizumab, was associated with prolonged OS, compared with docetaxel in the EGFR wild-type subgroup but not in the EGFR mutant subgroup [167]. ATLANTIC is a phase 2, open-label, single-arm trial concerning durvalumab as third-line or later treatment for advanced NSCLC. Although EGFR mutant NSCLC with PD-L1 expression of ≥ 25% had a higher response rate than the low expression group (12.2% vs. 3.6%), it still responded worse than the EGFR/ALK wild-type cohorts [168]. The WJOG8515L study is a randomized phase II trial comparing nivolumab with carboplatin and pemetrexed treatment in EGFR mutant NSCLC resistant to EGFR-TKIs due to mechanisms other than T790M. The PFS of nivolumab was shorter than chemotherapy (1.7–5.6 months), although OS was similar between the two arms [169].

The reasons for EGFR mutant NSCLC showed little benefits to ICIs are as following. EGFR activation induces CD73 expression, which can subsequently reduce the expression of interferon γ mRNA signature [170]. Meanwhile, high CD73 expression in NSCLC and other tumors is associated with low PD-L1 expression and densities of CD8+ tumor-infiltrating lymphocytes (TIL) [171]. Simultaneously, tumor mutational burden (TMB) is positively associated with ICI effectiveness and is lower in the EGFR mutant tumors compared with EGFR wild-type tumors (median 3.77 vs. 6.12 mutations/Mb, P < 0.0001) [172].

Retrospective studies or subgroup analyses revealed that the limited evidence indicates the characteristics of EGFR mutant tumor that can possibly benefit from ICIs:

(1) Tumors with inflammatory TME. In a study concerting nivolumab monotherapy in tumors with EGFR mutation, nivolumab responders had a significantly higher CD8+ TIL density and nonsynonymous mutation burden [173]. The PFS of anti-PD-1 agents in the EGFR mutant tumor was longer in patients with high PD-L1 expression and high TMB [172,174]. Although patients with EGFR mutant NSCLC tend to have lower TMB and PD-L1 expression [172], studies indicated that EGFR-TKI treatment can change the unfavorable TME. In a cohort of 138 patients with EGFR mutant NSCLC, the proportion of PD-L1 high expression (≥ 50%) and the TMB level both significantly increased after EGFR-TKI treatment. Moreover, in this cohort, patients with PD-L1 expression increasing from low to high after that EGFR-TKI treatment achieved a PFS of > 6 months [174]. In another cohort of 153 patients with EGFR mutant lung cancer, TMB also increased at resistance (median 3.42 vs. 6.56 mutations/Mb, P = 0.008) [172].

(2) Short PFS to EGFR-TKIs. A study including 58 patients with EGFR mutant NSCLC that received ICI treatment after EGFR-TKI treatment found that patients with shorter TKI-PFS (PFS of EGFR-TKIs was shorter than 10 months) had a significantly prolonged PFS and higher ORR of ICI treatment than those with longer TKI-PFS. Patients with shorter TKI-PFS harbored a higher proportion of CD8+ T cells and a lower ratio of M1 and M2-like macrophages [175].

(3) EGFR L858R mutation. A retrospective study reported that patients with EGFR L858R mutation showed similar clinical outcomes compared with the EGFR wild-type cohort in anti-PD-1/PD-L1 inhibitor treatment. However, the outcomes were poorer in patients with EGFR exon 19 deletion [176].

(4) T790M-negative patients. Clinical data indicated that the PFS of nivolumab after EGFR-TKIs treatment failure was longer for T790M-negative than T790M-positive patients (2.1 vs. 1.3 months, P = 0.099) [173].

For the driver mutation other than EGFR, the current National Comprehensive Cancer Network (NCCN) guidelines also recommended the use of target therapy as the first-line treatment for advanced NSCLC, such as ALK, ROS1, BRAF, NTRK1/2/3, MET ex14 skipping, and RET. After the disease progression, systematic therapy is recommended. By reviewing the IMMUNOTARGET registry and other published data considering ICIs in NSCLC with oncogenic driver alteration, we found that the ORR and PFS for NSCLC harboring the KRAS, BRAF, and c-MET alterations were comparable to non-mutant NSCLC in ICI registration trials, whereas the ORR in ALK, ROS-1 and RET fusion and NSCLC were relatively low. In the IMMUNOTARGET study, PD-L1 expression was relatively high in those ALK, ROS-1 and RET translocation cases [177,178]. Limited data showed variable ORRs of HER2, ROS1, and NTRK-altered NSCLCs, leaving an unclear efficacy of ICIs. Generally, patients with actionable tumor alternations show limited benefit form ICI monotherapy, combination of ICIs and chemotherapy and/or antiangiogenic agents is an option. New relevant biomarkers besides PD-L1 expression should be explored for these oncogenic addicted tumors.

Furthermore, the benefits to ICIs in KRAS mutant NSCLCs may vary depending on co-mutations. Co-occurring KRAS mutations and TP53 inactivation are associated with favorable responses to ICI monotherapy. Conversely, KRAS mutation co-occurred with liver kinase B1 (LKB1)/STK11 or Kelch-like ECH-associated protein 1 (KEAP1)-nuclear factor erythroid 2-related factor 2 (NRF2) pathway function loss will lead to immunosuppressed phenotypes, thus resulting in resistantance to PD-1/PD-L1 inhibitors [179181].

4.2 Potential mechanisms of low ICI efficacy in EGFR mutant NSCLC

EGFR mutations promote an immunosuppressive TME. First, EGFR mutant cancer downregulates CXC-chemokine ligand 10 (CXCL10) via phosphatidylinositol-3 kinase (PI3K)–AKT pathways, resulting in low infiltration of CD8+ T cells. Second, EGFR signaling upregulates the CC-chemokine ligand 22 (CCL22) expression through the JNK–JUN activation, thus recruiting the T-regulatory cells (Tregs). Treg cells can secrete IL-10, IL-35, and TGF-β that reduce antitumor immune responses mediated by natural killer (NK) cells and CD8+ T cells. Third, the activation of EGFR downstream pathways produces pro-tumoral cytokines and EGFR ligands that promote the macrophages 2 (M2) polarization of tumor-associated macrophages (TAMs), which are associated with suppressed cytotoxic T cell function. Fourth, CD39/CD73 are overexpressed in EGFR-mutated NSCLCs, which triggers the de-phosphorylation of adenosine triphosphate (ATP) to adenosine diphosphate (ADP), and subsequently results in adenosine monophosphate (AMP) and adenosine (ADO). ADO functions as an immunosuppressive mediator in the TME. The CD73-ADO axis promotes the Treg efficacy and myeloid-derived suppressor cell (MDSC). ADO combined with A2A adenosine receptor (A2AR) also inhibits T cell signal transduction by enhanced immunosuppressive activity of Treg cells and MDSCs [182184].

The immunosuppressive TME induced by EGFR mutation enables tumors to escape from antitumor immune responses. Thus, directly targeting EGFR or upregulating the tumor antigen presentation, inactivating immunosuppressive cells, and downregulating immunosuppressive molecules may lead to antitumor immunity in EGFR mutant tumors.

4.3 Strategies for ICI treatment in NSCLC with driver mutations

NSCLC with driver mutations generally showed limited ICI responses. Strategies for immunotherapy (IO) in NSCLC with driver mutations include the following.

4.3.1 IO + target therapy

A series of studies explored the combination of immunotherapy and target therapy [185187]. However, nearly all clinical trials were discontinued due to severe adverse events and failed to increase the efficacy compared with the target monotherapy. Combination of durvalumab and gefitinib failed to increase PFS and had higher hepatic toxicity than either agent alone [188]. Pembrolizumab plus gefitinib was also reported to have liver toxicity and failed to increase ORR [189]. Pembrolizumab or nivolumab plus erlotinib seems tolerable though still does not improve the ORR compared with monotherapy [189,190]. TATTON and CAURAL trial explored the combination use of osimertinib plus durvalumab. Both studies showed high incidence of interstitial lung disease (ILD) [185]. Immunotherapy in combination with first-generation seems prone to cause hepatic toxicity while third-generation EGFR-TKI with higher incidence of ILD. The mechanisms to explain the synergistic toxicity include that EGFR-TKIs might enhance T cell-mediated killing of tumor cells by promoting the MHC-I presentation [192]. In that case, EGFR-TKIs might also have immunomodulatory effects. EGFR signaling also has an impact on the microenvironment, suppressing immune-mediated anti-tumor responses via cytokines such as IL-6, TGF-β1, and progranulin [193]. The abnormality of these cytokines may also result in adverse effect.

4.3.2 IO + chemotherapy ± angiogenesis therapy

The IMpower150 study included 124 patients with EGFR mutant NSCLC. In this EGFR mutation cohort, the median PFS (10.2 vs. 6.9 months) and response duration (DOR, 11.1 vs. 4.7 months) were significantly improved in the atezolizumab plus bevacizumab plus carboplatin plus paclitaxel (ABCP) group versus the standard-of-care bevacizumab plus carboplatin plus paclitaxel in chemotherapy-naive patients. The difference was not significant when ABCP is compared with atezolizumab plus carboplatin plus paclitaxel group, indicating that the angiogenesis therapy is essential in the combination IO therapy in EGFR-mutant NSCLC [194]. ORIENT-31 (NCT03802240) compared sintilimab ± IBI305 plus chemotherapy (pemetrexed + cisplatin) for patients with EGFR mutant locally advanced or metastasis non-squamous NSCLC after the EGFR-TKI treatment failed. With a median follow-up time of 9.8 months, ORIENT-31 met its primary end-point of PFS with sintilimab + IBI305 + chemotherapy, achieving a median PFS of 6.9 months versus 4.3 months for chemotherapy alone (HR = 0.464, 95% CI 0.337–0.639) [195]. Two randomized phase 3 trails, Keynote-789 (NCT03515837) and CT25 (NCT03924050), were designed to assess the beneficial role of the addition of ICIs to chemotherapy in the EGFR-TKI-resistant setting. CheckMate722 (NCT02864251) evaluated the efficacy of nivolumab + chemotherapy and nivolumab + ipilimumab compared with chemotherapy in patients with EGFR mutation that progressed on the first-line EGFR-TKI therapy. These results have not been published.

Several early-stage studies showed promising antitumor activity of IO + chemotherapy in patients with NSCLC who progressed on EGFR-TKIs. A phase II study evaluated the efficacy of pembrolizumab combined with carboplatin and pemetrexed in recurrent EGFR/ALK + NSCLC, and results showed that IO + chemotherapy led to a response rate of 42% and a median OS of 22.2 months in EGFR mutant NSCLC; however, the combination had limited activity in seven patients with recurrent ALK-positive disease [196]. Similarly, in a phase II study that evaluated the efficacy of tislelizumab plus chemotherapy in patients with EGFR-mutated advanced non-squamous NSCLC who failed to EGFR-TKI therapies, the initial analysis showed that ORR and DCR were 59.4% (95%CI 40.6%–76.3%) and 90.6% (95%CI 75.0%–98.0%), respectively [197].

4.3.3 IO + angiogenesis therapy

In the phase I study that evaluated the efficacy and safety of TQB2450 (a PD-L1 inhibitor) combined with anlotinib in patients with advanced NSCLC after EGFR-TKI resistance, the results showed that the combination therapy was well tolerated with potential clinical antitumor activity, with a DCR rate of 77.8% and a median PFS of 9.1 months. Combined apatinib and camrelizumab in a phase II cohort study also exhibited encouraging antitumor activity and acceptable toxicity in EGFR mutant NSCLC after the TKI resistance, demonstrating an ORR of 18.6% and a median PFS of 2.8 months [198].

4.3.4 Emerging immunotherapies

NSCLC with driver mutations generally respond poorly to PD-1/PD-L1 inhibitors. The immunosuppressive TME mainly composed by Treg, MDSC, and M2-TAM is a limitation for the T cell-induced antitumor activity. To change the immunosuppressive TME, increasing evidences sustain the role of new additional inhibitory immune checkpoint molecules, such as T cell immunoglobulin and mucin-containing molecule 3 (TIM-3), lymphocyte activation gene-3 (LAG-3), and T cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT) to overcome the resistance to traditional ICIs [199201]. The CD39/CD73/adenosine signaling-mediated dysregulation of energy metabolism in TME is suggested as a potential mechanism involved in the immune escape process. Hence, there is a strong rationale of the combination therapy of ICIs with adenosine signaling inhibitors to overcome immunotherapy resistance in EGFR-mutated NSCLC [202,203]. Several clinical trials evaluating the efficacy of several A2AR inhibitors and anti-CD73 monoclonal antibodies for EGFR mutant NSCLC are in progress. Other novel immunotherapy approaches such as the T cell transfer therapy, TIL therapy, and chimeric antigen receptor-modified T (CAR-T) cell therapy, also have potency to control tumor growth in the presence of an appropriate immune context in NSCLC with driver mutations (Tab.2) [204,205].

5 New target therapies for NSCLC with driver mutations

Multiple novel oncogenic drivers have been identified in NSCLC. SHP2 (Src homology 2-containing phosphotyrosine phosphatase 2) plays an important role in regulating multiple cascade signaling pathways, including the Ras-Raf-MEK-ERK, PI3K-AKT, JAK-STAT, and PD-1/PD-L1 pathways. SHP2 inhibitors might be a promising drug that could synergistically sensitize the antitumor activity of target drugs in the oncogenic-addicted NSCLC [206]. To date, several small-molecule SHP2 inhibitors have advanced into clinical trials for mono- or combined therapy of cancers [207]. Fusions involving the neuregulin-1 (NRG1) gene result in ErbB pathway activation and therefore can be a candidate for targeted treatment [208]. Pan-HER tyrosine kinase inhibitor afatinib has reported the activity in NRG1 fusion NSCLC [208,209]. Another potential strategy is the use of HER3 inhibitors (GSK2849330) that showed the antitumor activity for CD74-NRG1 gene fusion tumor in a phase I trial [210].

Receptor tyrosine-protein kinase ERBB3 (HER3) is wildly expressed in NSCLC. HER3 overexpression is associated with metastatic progression and disease relapse in NSCLC. Increased HER3 expression has been observed in EGFR-resistant cell lines, indicating that HER3 overexpression contributed to drug resistance. HER3 antibody–drug conjugate (ADC) has shown clinical activity in EGFR TKI-resistant cancers, regardless of resistance mechanisms [211]. However, many new target agonists or inhibitors and ADCs with new antibodies or payloads are in clinical or preclinical studies that will considerably help the oncogene-driven personalized treatment of NSCLC (Tab.2).

6 Conclusions and perspectives

First, the discovery of oncogenic driver alterations and target therapy have brought significant clinical benefits and established an individualized treatment approach. The management of advanced NSCLC has shifted from a histology based on a biomarker-driven process.

Second, the treatment landscape of oncogenic-addicted NSCLC has become complex. On the one hand, more individualized treatment based on fine stratification has become the focus of research nowadays. The choice of optimal treatment strategy should consider gene subtype, concomitant mutation, dynamic gene alternation, and metastasis site. On the other hand, understanding primary and acquired resistance to targeted therapy provides an insight into the molecular evolution of tumor development. The recognition of resistant mechanisms is the basis to design new drugs or combinatorial therapeutic strategies.

Lastly, combination strategies require integration with immunotherapy, and the immunosuppressive microenvironment should be reversed to improve the sensitivity of ICIs by the drug combination. We should also be aware of the possible risk of combined toxicity and thereby explore the optimal timing and combined regimen of immunotherapy. Meanwhile, the interaction between driver mutations and immune-microenvironment is essential to uncover after drug resistance and to establish robust predictive biomarker for the NSCLC with driver mutations, in order to identify specific oncogenic driving patients with NSCLC who can benefit from immunotherapy.

References

[1]

Zhou C, Wu YL, Chen G, Feng J, Liu XQ, Wang C, Zhang S, Wang J, Zhou S, Ren S, Lu S, Zhang L, Hu C, Hu C, Luo Y, Chen L, Ye M, Huang J, Zhi X, Zhang Y, Xiu Q, Ma J, Zhang L, You C. Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol 2011; 12(8): 735–742

[2]

Sequist LV, Yang JCH, Yamamoto N, O’Byrne K, Hirsh V, Mok T, Geater SL, Orlov S, Tsai CM, Boyer M, Su WC, Bennouna J, Kato T, Gorbunova V, Lee KH, Shah R, Massey D, Zazulina V, Shahidi M, Schuler M. Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. J Clin Oncol 2013; 31(27): 3327–3334

[3]

Mok TS, Wu YL, Ahn MJ, Garassino MC, Kim HR, Ramalingam SS, Shepherd FA, He Y, Akamatsu H, Theelen WS, Lee CK, Sebastian M, Templeton A, Mann H, Marotti M, Ghiorghiu S, Papadimitrakopoulou VA; AURA3 Investigators. Osimertinib or platinum-pemetrexed in EGFR T790M-positive lung cancer. N Engl J Med 2017; 376(7): 629–640

[4]

Friboulet L, Li N, Katayama R, Lee CC, Gainor JF, Crystal AS, Michellys PY, Awad MM, Yanagitani N, Kim S, Pferdekamper AC, Li J, Kasibhatla S, Sun F, Sun X, Hua S, McNamara P, Mahmood S, Lockerman EL, Fujita N, Nishio M, Harris JL, Shaw AT, Engelman JA. The ALK inhibitor ceritinib overcomes crizotinib resistance in non-small cell lung cancer. Cancer Discov 2014; 4(6): 662–673

[5]

Gainor JF, Dardaei L, Yoda S, Friboulet L, Leshchiner I, Katayama R, Dagogo-Jack I, Gadgeel S, Schultz K, Singh M, Chin E, Parks M, Lee D, DiCecca RH, Lockerman E, Huynh T, Logan J, Ritterhouse LL, Le LP, Muniappan A, Digumarthy S, Channick C, Keyes C, Getz G, Dias-Santagata D, Heist RS, Lennerz J, Sequist LV, Benes CH, Iafrate AJ, Mino-Kenudson M, Engelman JA, Shaw AT. Molecular mechanisms of resistance to first- and second-generation ALK inhibitors in ALK-rearranged lung cancer. Cancer Discov 2016; 6(10): 1118–1133

[6]

Zhang S, Anjum R, Squillace R, Nadworny S, Zhou T, Keats J, Ning Y, Wardwell SD, Miller D, Song Y, Eichinger L, Moran L, Huang WS, Liu S, Zou D, Wang Y, Mohemmad Q, Jang HG, Ye E, Narasimhan N, Wang F, Miret J, Zhu X, Clackson T, Dalgarno D, Shakespeare WC, Rivera VM. The potent ALK inhibitor brigatinib (AP26113) overcomes mechanisms of resistance to first- and second-generation ALK inhibitors in preclinical models. Clin Cancer Res 2016; 22(22): 5527–5538

[7]

Peters S, Camidge DR, Shaw AT, Gadgeel S, Ahn JS, Kim DW, Ou SI, Pérol M, Dziadziuszko R, Rosell R, Zeaiter A, Mitry E, Golding S, Balas B, Noe J, Morcos PN, Mok T; ALEX Trial Investigators. Alectinib versus crizotinib in untreated ALK-positive non-small-cell lung cancer. N Engl J Med 2017; 377(9): 829–838

[8]

Shaw AT, Bauer TM, de Marinis F, Felip E, Goto Y, Liu G, Mazieres J, Kim DW, Mok T, Polli A, Thurm H, Calella AM, Peltz G, Solomon BJ; CROWN Trial Investigators. First-line lorlatinib or crizotinib in advanced ALK-positive lung cancer. N Engl J Med 2020; 383(21): 2018–2029

[9]

Camidge DR, Kim HR, Ahn MJ, Yang JCH, Han JY, Hochmair MJ, Lee KH, Delmonte A, Garcia Campelo MR, Kim DW, Griesinger F, Felip E, Califano R, Spira AI, Gettinger SN, Tiseo M, Lin HM, Liu Y, Vranceanu F, Niu H, Zhang P, Popat S. Brigatinib versus crizotinib in ALK inhibitor-naive advanced ALK-positive NSCLC: final results of phase 3 ALTA-1L trial. J Thorac Oncol 2021; 16(12): 2091–2108

[10]

Solomon BJ, Mok T, Kim DW, Wu YL, Nakagawa K, Mekhail T, Felip E, Cappuzzo F, Paolini J, Usari T, Iyer S, Reisman A, Wilner KD, Tursi J, Blackhall F; PROFILE 1014 Investigators. First-line crizotinib versus chemotherapy in ALK-positive lung cancer. N Engl J Med 2014; 371(23): 2167–2177

[11]

Shaw AT, Ou SHI, Bang YJ, Camidge DR, Solomon BJ, Salgia R, Riely GJ, Varella-Garcia M, Shapiro GI, Costa DB, Doebele RC, Le LP, Zheng Z, Tan W, Stephenson P, Shreeve SM, Tye LM, Christensen JG, Wilner KD, Clark JW, Iafrate AJ. Crizotinib in ROS1-rearranged non-small-cell lung cancer. N Engl J Med 2014; 371(21): 1963–1971

[12]

Planchard D, Besse B, Groen HJM, Souquet PJ, Quoix E, Baik CS, Barlesi F, Kim TM, Mazieres J, Novello S, Rigas JR, Upalawanna A, D’Amelio AM Jr, Zhang P, Mookerjee B, Johnson BE. Dabrafenib plus trametinib in patients with previously treated BRAF (V600E)-mutant metastatic non-small cell lung cancer: an open-label, multicentre phase 2 trial. Lancet Oncol 2016; 17(7): 984–993

[13]

Haratake N, Seto T. NTRK fusion-positive non-small-cell lung cancer: the diagnosis and targeted therapy. Clin Lung Cancer 2021; 22(1): 1–5

[14]

Uprety D, Adjei AA. KRAS: from undruggable to a druggable cancer target. Cancer Treat Rev 2020; 89: 102070

[15]

da Cunha Santos G, Shepherd FA, Tsao MS. EGFR mutations and lung cancer. Annu Rev Pathol 2011; 6(1): 49–69

[16]

He Q, Xin P, Zhang M, Jiang S, Zhang J, Zhong S, Liu Y, Guo M, Chen X, Xia X, Pan Z, Guo C, Cai X, Liang W, He J. The impact of epidermal growth factor receptor mutations on the prognosis of resected non-small cell lung cancer: a meta-analysis of literatures. Transl Lung Cancer Res 2019; 8(2): 124–134

[17]

Lee CK, Wu YL, Ding PN, Lord SJ, Inoue A, Zhou C, Mitsudomi T, Rosell R, Pavlakis N, Links M, Gebski V, Gralla RJ, Yang JC. Impact of specific epidermal growth factor receptor (EGFR) mutations and clinical characteristics on outcomes after treatment with EGFR tyrosine kinase inhibitors versus chemotherapy in EGFR-mutant lung cancer: a meta-analysis. J Clin Oncol 2015; 33(17): 1958–1965

[18]

Yang JCH, Wu YL, Schuler M, Sebastian M, Popat S, Yamamoto N, Zhou C, Hu CP, O’Byrne K, Feng J, Lu S, Huang Y, Geater SL, Lee KY, Tsai CM, Gorbunova V, Hirsh V, Bennouna J, Orlov S, Mok T, Boyer M, Su WC, Lee KH, Kato T, Massey D, Shahidi M, Zazulina V, Sequist LV. Afatinib versus cisplatin-based chemotherapy for EGFR mutation-positive lung adenocarcinoma (LUX-Lung 3 and LUX-Lung 6): analysis of overall survival data from two randomised, phase 3 trials. Lancet Oncol 2015; 16(2): 141–151

[19]

Ramalingam SS, Vansteenkiste J, Planchard D, Cho BC, Gray JE, Ohe Y, Zhou C, Reungwetwattana T, Cheng Y, Chewaskulyong B, Shah R, Cobo M, Lee KH, Cheema P, Tiseo M, John T, Lin MC, Imamura F, Kurata T, Todd A, Hodge R, Saggese M, Rukazenkov Y, Soria JC; FLAURA Investigators. Overall survival with osimertinib in untreated, EGFR-mutated advanced NSCLC. N Engl J Med 2020; 382(1): 41–50

[20]

Saito H, Fukuhara T, Furuya N, Watanabe K, Sugawara S, Iwasawa S, Tsunezuka Y, Yamaguchi O, Okada M, Yoshimori K, Nakachi I, Gemma A, Azuma K, Kurimoto F, Tsubata Y, Fujita Y, Nagashima H, Asai G, Watanabe S, Miyazaki M, Hagiwara K, Nukiwa T, Morita S, Kobayashi K, Maemondo M. 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(5): 625–635

[21]

Zhou Q, Xu CR, Cheng Y, Liu YP, Chen GY, Cui JW, Yang N, Song Y, Li XL, Lu S, Zhou JY, Ma ZY, Yu SY, Huang C, Shu YQ, Wang Z, Yang JJ, Tu HY, Zhong WZ, Wu YL. Bevacizumab plus erlotinib in Chinese patients with untreated, EGFR-mutated, advanced NSCLC (ARTEMIS-CTONG1509): a multicenter phase 3 study. Cancer Cell 2021; 39(9): 1279–1291.e3

[22]

Li X, Zhang L, Jiang D, Wang Y, Zang A, Ding C, Zhao M, Su W, Zhang Y, Zhong D, Wu J, Zhang C, An G, Hu X, Cheng G, Wang H, Li Y, He X, Liu J, Liang L, Ding L, Mao L, Zhang S. Routine-dose and high-dose icotinib in patients with advanced non-small cell lung cancer harboring EGFR exon 21-L858R mutation: the randomized, phase II, INCREASE trial. Clin Cancer Res 2020; 26(13): 3162–3171

[23]

Passaro A, Mok T, Peters S, Popat S, Ahn MJ, de Marinis F. Recent advances on the role of EGFR tyrosine kinase inhibitors in the management of NSCLC with uncommon, non exon 20 insertions, EGFR mutations. J Thorac Oncol 2021; 16(5): 764–773

[24]

Yang JCH, Sequist LV, Geater SL, Tsai CM, Mok TS, Schuler M, Yamamoto N, Yu CJ, Ou SH, Zhou C, Massey D, Zazulina V, Wu YL. Clinical activity of afatinib in patients with advanced non-small-cell lung cancer harbouring uncommon EGFR mutations: a combined post-hoc analysis of LUX-Lung 2, LUX-Lung 3, and LUX-Lung 6. Lancet Oncol 2015; 16(7): 830–838

[25]

Masood A, Kancha RK, Subramanian J. Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors in non-small cell lung cancer harboring uncommon EGFR mutations: focus on afatinib. Semin Oncol 2019; 46(3): 271–283

[26]

Vyse S, Huang PH. Amivantamab for the treatment of EGFR exon 20 insertion mutant non-small cell lung cancer. Expert Rev Anticancer Ther 2022; 22(1): 3–16

[27]

Park K, Haura EB, Leighl NB, Mitchell P, Shu CA, Girard N, Viteri S, Han JY, Kim SW, Lee CK, Sabari JK, Spira AI, Yang TY, Kim DW, Lee KH, Sanborn RE, Trigo J, Goto K, Lee JS, Yang JC, Govindan R, Bauml JM, Garrido P, Krebs MG, Reckamp KL, Xie J, Curtin JC, Haddish-Berhane N, Roshak A, Millington D, Lorenzini P, Thayu M, Knoblauch RE, Cho BC. Amivantamab in EGFR exon 20 insertion-mutated non-small-cell lung cancer progressing on platinum chemotherapy: initial results from the CHRYSALIS phase I study. J Clin Oncol 2021; 39(30): 3391–3402

[28]

Yang S, Mao S, Li X, Zhao C, Liu Q, Yu X, Wang Y, Liu Y, Pan Y, Wang C, Gao G, Li W, Xiong A, Chen B, Sun H, He Y, Wu F, Chen X, Su C, Ren S, Zhou C. Uncommon EGFR mutations associate with lower incidence of T790M mutation after EGFR-TKI treatment in patients with advanced NSCLC. Lung Cancer 2020; 139: 133–139

[29]

Wang H, Zhang M, Tang W, Ma J, Wei B, Niu Y, Zhang G, Li P, Yan X, Ma Z. Mutation abundance affects the therapeutic efficacy of EGFR-TKI in patients with advanced lung adenocarcinoma: a retrospective analysis. Cancer Biol Ther 2018; 19(8): 687–694

[30]

Wang X, Liu Y, Meng Z, Wu Y, Wang S, Jin G, Qin Y, Wang F, Wang J, Zhou H, Su X, Fu X, Wang X, Shi X, Wen Z, Jia X, Qin Q, Gao Y, Guo W, Lu S. Plasma EGFR mutation abundance affects clinical response to first-line EGFR-TKIs in patients with advanced non-small cell lung cancer. Ann Transl Med 2021; 9(8): 635

[31]

Pan G, Chen K, Yu X, Sheng J, Fan Y. The correlation between the abundance of EGFR T790M mutation and osimertinib response in advanced non-small cell lung cancer. Transl Cancer Res 2021; 10(6): 2895–2905

[32]

Furugaki K, Harada N, Yoshimura Y. Sensitivity of eight types of ALK fusion variant to alectinib in ALK-transformed cells. Anticancer Drugs 2022; 33(2): 124–131

[33]

Wang S, Luo R, Shi Y, Han X. The impact of the ALK fusion variant on clinical outcomes in EML4-ALK patients with NSCLC: a systematic review and meta-analysis. Future Oncol 2022; 18(3): 385–402

[34]

Zhang SS, Nagasaka M, Zhu VW, Ou SI. Going beneath the tip of the iceberg. Identifying and understanding EML4-ALK variants and TP53 mutations to optimize treatment of ALK fusion positive (ALK+) NSCLC. Lung Cancer 2021; 158: 126–136

[35]

Yaeger R, Corcoran RB. Targeting alterations in the RAF-MEK pathway. Cancer Discov 2019; 9(3): 329–341

[36]

Dankner M, Lajoie M, Moldoveanu D, Nguyen TT, Savage P, Rajkumar S, Huang X, Lvova M, Protopopov A, Vuzman D, Hogg D, Park M, Guiot MC, Petrecca K, Mihalcioiu C, Watson IR, Siegel PM, Rose AAN. Dual MAPK inhibition is an effective therapeutic strategy for a subset of class II BRAF mutant melanomas. Clin Cancer Res 2018; 24(24): 6483–6494

[37]

Negrao MV, Raymond VM, Lanman RB, Robichaux JP, He J, Nilsson MB, Ng PKS, Amador BE, Roarty EB, Nagy RJ, Banks KC, Zhu VW, Ng C, Chae YK, Clarke JM, Crawford JA, Meric-Bernstam F, Ignatius Ou SH, Gandara DR, Heymach JV, Bivona TG, McCoach CE. Molecular landscape of BRAF-mutant NSCLC reveals an association between clonality and driver mutations and identifies targetable non-V600 driver mutations. J Thorac Oncol 2020; 15(10): 1611–1623

[38]

Tan AC, Seet AOL, Lai GGY, Lim TH, Lim AST, Tan GS, Takano A, Tai DWM, Tan TJY, Lam JYC, Ng MCH, Tan WL, Ang MK, Kanesvaran R, Ng QS, Jain A, Rajasekaran T, Lim WT, Tan EH, Lim TKH, Tan DSW. Molecular characterization and clinical outcomes in RET-rearranged NSCLC. J Thorac Oncol 2020; 15(12): 1928–1934

[39]

Feng J, Li Y, Wei B, Guo L, Li W, Xia Q, Zhao C, Zheng J, Zhao J, Sun R, Guo Y, Brcic L, Hakozaki T, Ying J, Ma J. Clinicopathologic characteristics and diagnostic methods of RET rearrangement in Chinese non-small cell lung cancer patients. Transl Lung Cancer Res 2022; 11(4): 617–631

[40]

ArtsimovichNGNastoiashchaiaNNLymar’ NPKostrovaAAOsokinaLI. Effect of antibiotics on hematologic indices and enzyme activity of blood lymphocytes in mice. Gematol Transfuziol 1987; 32(4): 58–62 (in Russian)

[41]

Garon EB, Brodrick P. Targeted therapy approaches for MET abnormalities in non-small cell lung cancer. Drugs 2021; 81(5): 547–554

[42]

Coleman N, Harbery A, Heuss S, Vivanco I, Popat S. Targeting un-MET needs in advanced non-small cell lung cancer. Lung Cancer 2022; 164: 56–68

[43]

Drusbosky LM, Dawar R, Rodriguez E, Ikpeazu CV. Therapeutic strategies in METex14 skipping mutated non-small cell lung cancer. J Hematol Oncol 2021; 14(1): 129

[44]

Liu Q, Yu S, Zhao W, Qin S, Chu Q, Wu K. EGFR-TKIs resistance via EGFR-independent signaling pathways. Mol Cancer 2018; 17(1): 53

[45]

Hong S, Gao F, Fu S, Wang Y, Fang W, Huang Y, Zhang L. Concomitant genetic alterations with response to treatment and epidermal growth factor receptor tyrosine kinase inhibitors in patients with EGFR-mutant advanced non-small cell lung cancer. JAMA Oncol 2018; 4(5): 739–742

[46]

Zhong J, Li L, Wang Z, Bai H, Gai F, Duan J, Zhao J, Zhuo M, Wang Y, Wang S, Zang W, Wu M, An T, Rao G, Zhu G, Wang J. Potential resistance mechanisms revealed by targeted sequencing from lung adenocarcinoma patients with primary resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs). J Thorac Oncol 2017; 12(12): 1766–1778

[47]

Wang Z, Cheng Y, An T, Gao H, Wang K, Zhou Q, Hu Y, Song Y, Ding C, Peng F, Liang L, Hu Y, Huang C, Zhou C, Shi Y, Zhang L, Ye X, Zhang M, Chuai S, Zhu G, Hu J, Wu YL, Wang J. Detection of EGFR mutations in plasma circulating tumour DNA as a selection criterion for first-line gefitinib treatment in patients with advanced lung adenocarcinoma (BENEFIT): a phase 2, single-arm, multicentre clinical trial. Lancet Respir Med 2018; 6(9): 681–690

[48]

Duan J, Xu J, Wang Z, Bai H, Cheng Y, An T, Gao H, Wang K, Zhou Q, Hu Y, Song Y, Ding C, Peng F, Liang L, Hu Y, Huang C, Zhou C, Shi Y, Han J, Wang D, Tian Y, Yang Z, Zhang L, Chuai S, Ye J, Zhu G, Zhao J, Wu YL, Wang J. Refined stratification based on baseline concomitant mutations and longitudinal circulating tumor DNA monitoring in advanced EGFR-mutant lung adenocarcinoma under gefitinib treatment. J Thorac Oncol 2020; 15(12): 1857–1870

[49]

La Monica S, Minari R, Cretella D, Flammini L, Fumarola C, Bonelli M, Cavazzoni A, Digiacomo G, Galetti M, Madeddu D, Falco A, Lagrasta CA, Squadrilli A, Barocelli E, Romanel A, Quaini F, Petronini PG, Tiseo M, Alfieri R. Third generation EGFR inhibitor osimertinib combined with pemetrexed or cisplatin exerts long-lasting anti-tumor effect in EGFR-mutated pre-clinical models of NSCLC. J Exp Clin Cancer Res 2019; 38(1): 222

[50]

Planchard D, Feng PH, Karaseva N, Kim SW, Kim TM, Lee CK, Poltoratskiy A, Yanagitani N, Marshall R, Huang X, Howarth P, Jänne PA, Kobayashi K. 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(5): 100271

[51]

Hosomi Y, Morita S, Sugawara S, Kato T, Fukuhara T, Gemma A, Takahashi K, Fujita Y, Harada T, Minato K, Takamura K, Hagiwara K, Kobayashi K, Nukiwa T, Inoue A; North-East Japan Study Group. 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(2): 115–123

[52]

Hsu WH, Yang JCH, Mok TS, Loong HH. Overview of current systemic management of EGFR-mutant NSCLC. Ann Oncol 2018; 29(suppl_1): i3–i9

[53]

Leonetti A, Sharma S, Minari R, Perego P, Giovannetti E, Tiseo M. Resistance mechanisms to osimertinib in EGFR-mutated non-small cell lung cancer. Br J Cancer 2019; 121(9): 725–737

[54]

He J, Huang Z, Han L, Gong Y, Xie C. Mechanisms and management of 3rd-generation EGFR-TKI resistance in advanced non-small cell lung cancer (Review). Int J Oncol 2021; 59(5): 90

[55]

Cho BC, Felip E, Hayashi H, Thomas M, Lu S, Besse B, Sun T, Martinez M, Sethi SN, Shreeve SM, Spira AI. MARIPOSA: phase 3 study of first-line amivantamab + lazertinib versus osimertinib in EGFR-mutant non-small-cell lung cancer. Future Oncol 2022; 18(6): 639–647

[56]

WuDLiJ YaoMHZheng YHFengCYYangYH. Clinicopathological significance in non-small cell lung cancer with mutations and co-mutations of EGFR, ALK and ROS1. Chin J Pathol (Zhonghua Bing Li Xue Za Zhi) 2021; 50(3): 251–253 (in Chinese)

[57]

Yang X, Zhong J, Yu Z, Zhuo M, Zhang M, Chen R, Xia X, Zhao J. Genetic and treatment profiles of patients with concurrent epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) mutations. BMC Cancer 2021; 21(1): 1107

[58]

Fan J, Dai X, Wang Z, Huang B, Shi H, Luo D, Zhang J, Cai W, Nie X, Hirsch FR. Concomitant EGFR mutation and EML4-ALK rearrangement in lung adenocarcinoma is more frequent in multifocal lesions. Clin Lung Cancer 2019; 20(4): e517–e530

[59]

Zhu YJ, Qu X, Zhan DD, Chen HH, Li HP, Liu LR, Chen X, Liu YH, Li Y, Bai JP, Ye S, Zhang HB. Specific gene co-variation acts better than number of concomitant altered genes in predicting EGFR-TKI efficacy in non-small-cell lung cancer. Clin Lung Cancer 2021; 22(1): e98–e111

[60]

Skoulidis F, Heymach JV. Co-occurring genomic alterations in non-small-cell lung cancer biology and therapy. Nat Rev Cancer 2019; 19(9): 495–509

[61]

Lou NN, Zhang XC, Chen HJ, Zhou Q, Yan LX, Xie Z, Su J, Chen ZH, Tu HY, Yan HH, Wang Z, Xu CR, Jiang BY, Wang BC, Bai XY, Zhong WZ, Wu YL, Yang JJ. Clinical outcomes of advanced non-small-cell lung cancer patients with EGFR mutation, ALK rearrangement and EGFR/ALK co-alterations. Oncotarget 2016; 7(40): 65185–65195

[62]

Kunimasa K, Hirotsu Y, Kukita Y, Ueda Y, Sato Y, Kimura M, Otsuka T, Hamamoto Y, Tamiya M, Inoue T, Kawamura T, Nishino K, Amemiya K, Goto T, Mochizuki H, Honma K, Omata M, Kumagai T. EML4-ALK fusion variant.3 and co-occurrent PIK3CA E542K mutation exhibiting primary resistance to three generations of ALK inhibitors. Cancer Genet 2021; 256-257: 131–135

[63]

Lu C, Dong XR, Zhao J, Zhang XC, Chen HJ, Zhou Q, Tu HY, Ai XH, Chen XF, An GL, Bai J, Shan JL, Wang YN, Yang SY, Liu X, Zhuang W, Wu HT, Zhu B, Xia XF, Chen RR, Gu DJ, Xu HM, Wu YL, Yang JJ. Association of genetic and immuno-characteristics with clinical outcomes in patients with RET-rearranged non-small cell lung cancer: a retrospective multicenter study. J Hematol Oncol 2020; 13(1): 37

[64]

Skoulidis F, Li BT, Dy GK, Price TJ, Falchook GS, Wolf J, Italiano A, Schuler M, Borghaei H, Barlesi F, Kato T, Curioni-Fontecedro A, Sacher A, Spira A, Ramalingam SS, Takahashi T, Besse B, Anderson A, Ang A, Tran Q, Mather O, Henary H, Ngarmchamnanrith G, Friberg G, Velcheti V, Govindan R. Sotorasib for lung cancers with KRAS p. G12C mutation. N Engl J Med 2021; 384(25): 2371–2381

[65]

Moiseenko FV, Volkov NM, Zhabina AS, Stepanova ML, Rysev NA, Klimenko VV, Myslik AV, Artemieva EV, Egorenkov VV, Abduloeva NH, Ivantsov AO, Kuligina ES, Imyanitov EN, Moiseyenko VM. Monitoring of the presence of EGFR-mutated DNA during EGFR-targeted therapy may assist in the prediction of treatment outcome. Cancer Treat Res Commun 2022; 31: 100524

[66]

Buder A, Hochmair MJ, Setinek U, Pirker R, Filipits M. EGFR mutation tracking predicts survival in advanced EGFR-mutated non-small cell lung cancer patients treated with osimertinib. Transl Lung Cancer Res 2020; 9(2): 239–245

[67]

Provencio M, Serna-Blasco R, Franco F, Calvo V, Royuela A, Auglytė M, Sánchez-Hernández A, de Julián Campayo M, García-Girón C, Dómine M, Blasco A, Sánchez JM, Oramas J, Bosch-Barrera J, Sala, Sereno M, Ortega AL, Chara L, Hernández B, Padilla A, Coves J, Blanco R, Balsalobre J, Mielgo X, Bueno C, Jantus-Lewintre E, Molina-Vila, Romero A. Analysis of circulating tumour DNA to identify patients with epidermal growth factor receptor-positive non-small cell lung cancer who might benefit from sequential tyrosine kinase inhibitor treatment. Eur J Cancer 2021; 149: 61–72

[68]

Wang W, Sun X, Hui Z. Treatment optimization for brain metastasis from anaplastic lymphoma kinase rearrangement non-small-cell lung cancer. Oncol Res Treat 2019; 42(11): 599–606

[69]

Mazzola R, Jereczek-Fossa BA, Franceschini D, Tubin S, Filippi AR, Tolia M, Lancia A, Minniti G, Corradini S, Arcangeli S, Scorsetti M, Alongi F. Oligometastasis and local ablation in the era of systemic targeted and immunotherapy. Radiat Oncol 2020; 15(1): 92

[70]

Gomez DR, Blumenschein GR Jr, Lee JJ, Hernandez M, Ye R, Camidge DR, Doebele RC, Skoulidis F, Gaspar LE, Gibbons DL, Karam JA, Kavanagh BD, Tang C, Komaki R, Louie AV, Palma DA, Tsao AS, Sepesi B, William WN, Zhang J, Shi Q, Wang XS, Swisher SG, Heymach JV. Local consolidative therapy versus maintenance therapy or observation for patients with oligometastatic non-small-cell lung cancer without progression after first-line systemic therapy: a multicentre, randomised, controlled, phase 2 study. Lancet Oncol 2016; 17(12): 1672–1682

[71]

Gomez DR, Tang C, Zhang J, Blumenschein GR Jr, Hernandez M, Lee JJ, Ye R, Palma DA, Louie AV, Camidge DR, Doebele RC, Skoulidis F, Gaspar LE, Welsh JW, Gibbons DL, Karam JA, Kavanagh BD, Tsao AS, Sepesi B, Swisher SG, Heymach JV. Local consolidative therapy vs. maintenance therapy or observation for patients with oligometastatic non-small-cell lung cancer: long-term results of a multi-institutional, phase II, randomized study. J Clin Oncol 2019; 37(18): 1558–1565

[72]

Weickhardt AJ, Scheier B, Burke JM, Gan G, Lu X, Bunn PA Jr, Aisner DL, Gaspar LE, Kavanagh BD, Doebele RC, Camidge DR. Local ablative therapy of oligoprogressive disease prolongs disease control by tyrosine kinase inhibitors in oncogene-addicted non-small-cell lung cancer. J Thorac Oncol 2012; 7(12): 1807–1814

[73]

Qiu B, Liang Y, Li Q, Liu G, Wang F, Chen Z, Liu M, Zhao M, Liu H. Local therapy for oligoprogressive disease in patients with advanced stage non-small-cell lung cancer harboring epidermal growth factor receptor mutation. Clin Lung Cancer 2017; 18(6): e369–e373

[74]

Weir BA, Woo MS, Getz G, Perner S, Ding L, Beroukhim R, Lin WM, Province MA, Kraja A, Johnson LA, Shah K, Sato M, Thomas RK, Barletta JA, Borecki IB, Broderick S, Chang AC, Chiang DY, Chirieac LR, Cho J, Fujii Y, Gazdar AF, Giordano T, Greulich H, Hanna M, Johnson BE, Kris MG, Lash A, Lin L, Lindeman N, Mardis ER, McPherson JD, Minna JD, Morgan MB, Nadel M, Orringer MB, Osborne JR, Ozenberger B, Ramos AH, Robinson J, Roth JA, Rusch V, Sasaki H, Shepherd F, Sougnez C, Spitz MR, Tsao MS, Twomey D, Verhaak RG, Weinstock GM, Wheeler DA, Winckler W, Yoshizawa A, Yu S, Zakowski MF, Zhang Q, Beer DG, Wistuba II, Watson MA, Garraway LA, Ladanyi M, Travis WD, Pao W, Rubin MA, Gabriel SB, Gibbs RA, Varmus HE, Wilson RK, Lander ES, Meyerson M. Characterizing the cancer genome in lung adenocarcinoma. Nature 2007; 450(7171): 893–898

[75]

Ni X, Zhuo M, Su Z, Duan J, Gao Y, Wang Z, Zong C, Bai H, Chapman AR, Zhao J, Xu L, An T, Ma Q, Wang Y, Wu M, Sun Y, Wang S, Li Z, Yang X, Yong J, Su XD, Lu Y, Bai F, Xie XS, Wang J. Reproducible copy number variation patterns among single circulating tumor cells of lung cancer patients. Proc Natl Acad Sci USA 2013; 110(52): 21083–21088

[76]

Beaubier N, Bontrager M, Huether R, Igartua C, Lau D, Tell R, Bobe AM, Bush S, Chang AL, Hoskinson DC, Khan AA, Kudalkar E, Leibowitz BD, Lozachmeur A, Michuda J, Parsons J, Perera JF, Salahudeen A, Shah KP, Taxter T, Zhu W, White KP. Integrated genomic profiling expands clinical options for patients with cancer. Nat Biotechnol 2019; 37(11): 1351–1360

[77]

Testa U, Pelosi E, Castelli G. Molecular charcterization of lung adenocarcinoma combining whole exome sequencing, copy number analysis and gene expression profiling. Expert Rev Mol Diagn 2022; 22(1): 77–100

[78]

Chen YJ, Roumeliotis TI, Chang YH, Chen CT, Han CL, Lin MH, Chen HW, Chang GC, Chang YL, Wu CT, Lin MW, Hsieh MS, Wang YT, Chen YR, Jonassen I, Ghavidel FZ, Lin ZS, Lin KT, Chen CW, Sheu PY, Hung CT, Huang KC, Yang HC, Lin PY, Yen TC, Lin YW, Wang JH, Raghav L, Lin CY, Chen YS, Wu PS, Lai CT, Weng SH, Su KY, Chang WH, Tsai PY, Robles AI, Rodriguez H, Hsiao YJ, Chang WH, Sung TY, Chen JS, Yu SL, Choudhary JS, Chen HY, Yang PC, Chen YJ. Proteogenomics of non-smoking lung cancer in East Asia delineates molecular signatures of pathogenesis and progression. Cell 2020; 182(1): 226–244.e17

[79]

Bivona TG, Doebele RC. A framework for understanding and targeting residual disease in oncogene-driven solid cancers. Nat Med 2016; 22(5): 472–478

[80]

Lee JK, Shin JY, Kim S, Lee S, Park C, Kim JY, Koh Y, Keam B, Min HS, Kim TM, Jeon YK, Kim DW, Chung DH, Heo DS, Lee SH, Kim JI. Primary resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) in patients with non-small-cell lung cancer harboring TKI-sensitive EGFR mutations: an exploratory study. Ann Oncol 2013; 24(8): 2080–2087

[81]

Jackman D, Pao W, Riely GJ, Engelman JA, Kris MG, Jänne PA, Lynch T, Johnson BE, Miller VA. Clinical definition of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer. J Clin Oncol 2010; 28(2): 357–360

[82]

Yang JJ, Chen HJ, Yan HH, Zhang XC, Zhou Q, Su J, Wang Z, Xu CR, Huang YS, Wang BC, Yang XN, Zhong WZ, Nie Q, Liao RQ, Jiang BY, Dong S, Wu YL. Clinical modes of EGFR tyrosine kinase inhibitor failure and subsequent management in advanced non-small cell lung cancer. Lung Cancer 2013; 79(1): 33–39

[83]

Yang SR, Schultheis AM, Yu H, Mandelker D, Ladanyi M, Büttner R. Precision medicine in non-small cell lung cancer: current applications and future directions. Semin Cancer Biol 2022; 84: 184–198

[84]

Robichaux JP, Le X, Vijayan RSK, Hicks JK, Heeke S, Elamin YY, Lin HY, Udagawa H, Skoulidis F, Tran H, Varghese S, He J, Zhang F, Nilsson MB, Hu L, Poteete A, Rinsurongkawong W, Zhang X, Ren C, Liu X, Hong L, Zhang J, Diao L, Madison R, Schrock AB, Saam J, Raymond V, Fang B, Wang J, Ha MJ, Cross JB, Gray JE, Heymach JV. Structure-based classification predicts drug response in EGFR-mutant NSCLC. Nature 2021; 597(7878): 732–737

[85]

Passaro A, Jänne PA, Mok T, Peters S. Overcoming therapy resistance in EGFR-mutant lung cancer. Nat Can 2021; 2(4): 377–391

[86]

Shah MP, Neal JW. Targeting acquired and intrinsic resistance mechanisms in epidermal growth factor receptor mutant non-small-cell lung cancer. Drugs 2022; 82(6): 649–662

[87]

Choi YL, Soda M, Yamashita Y, Ueno T, Takashima J, Nakajima T, Yatabe Y, Takeuchi K, Hamada T, Haruta H, Ishikawa Y, Kimura H, Mitsudomi T, Tanio Y, Mano H; ALK Lung Cancer Study Group. EML4-ALK mutations in lung cancer that confer resistance to ALK inhibitors. N Engl J Med 2010; 363(18): 1734–1739

[88]

McCoach CE, Le AT, Aisner D, Gowan K, Jones KL, Merrick D, Bunn PA, Purcell WT, Varella-Garcia M, Camidge DR, Doebele RC. Resistance mechanisms to targeted therapies in ROS1+ and ALK+ non-small cell lung cancer. J Clin Oncol 2016; 34(15 suppl): 9065

[89]

Lin JJ, Liu SV, McCoach CE, Zhu VW, Tan AC, Yoda S, Peterson J, Do A, Prutisto-Chang K, Dagogo-Jack I, Sequist LV, Wirth LJ, Lennerz JK, Hata AN, Mino-Kenudson M, Nardi V, Ou SI, Tan DS, Gainor JF. Mechanisms of resistance to selective RET tyrosine kinase inhibitors in RET fusion-positive non-small-cell lung cancer. Ann Oncol 2020; 31(12): 1725–1733

[90]

Tanaka N, Lin JJ, Li C, Ryan MB, Zhang J, Kiedrowski LA, Michel AG, Syed MU, Fella KA, Sakhi M, Baiev I, Juric D, Gainor JF, Klempner SJ, Lennerz JK, Siravegna G, Bar-Peled L, Hata AN, Heist RS, Corcoran RB. Clinical acquired resistance to KRASG12C inhibition through a novel KRAS switch-II pocket mutation and polyclonal alterations converging on RAS-MAPK reactivation. Cancer Discov 2021; 11(8): 1913–1922

[91]

Awad MM, Liu S, Rybkin II, Arbour KC, Dilly J, Zhu VW, Johnson ML, Heist RS, Patil T, Riely GJ, Jacobson JO, Yang X, Persky NS, Root DE, Lowder KE, Feng H, Zhang SS, Haigis KM, Hung YP, Sholl LM, Wolpin BM, Wiese J, Christiansen J, Lee J, Schrock AB, Lim LP, Garg K, Li M, Engstrom LD, Waters L, Lawson JD, Olson P, Lito P, Ou SI, Christensen JG, Jänne PA, Aguirre AJ. Acquired resistance to KRASG12C inhibition in cancer. N Engl J Med 2021; 384(25): 2382–2393

[92]

Koga T, Suda K, Fujino T, Ohara S, Hamada A, Nishino M, Chiba M, Shimoji M, Takemoto T, Arita T, Gmachl M, Hofmann MH, Soh J, Mitsudomi T. KRAS secondary mutations that confer acquired resistance to KRAS G12C inhibitors, sotorasib and adagrasib, and overcoming strategies: insights from in vitro experiments. J Thorac Oncol 2021; 16(8): 1321–1332

[93]

Kim TM, Song A, Kim DW, Kim S, Ahn YO, Keam B, Jeon YK, Lee SH, Chung DH, Heo DS. Mechanisms of acquired resistance to AZD9291: a mutation-selective, irreversible EGFR inhibitor. J Thorac Oncol 2015; 10(12): 1736–1744

[94]

Doebele RC, Pilling AB, Aisner DL, Kutateladze TG, Le AT, Weickhardt AJ, Kondo KL, Linderman DJ, Heasley LE, Franklin WA, Varella-Garcia M, Camidge DR. Mechanisms of resistance to crizotinib in patients with ALK gene rearranged non-small cell lung cancer. Clin Cancer Res 2012; 18(5): 1472–1482

[95]

Hua G, Zhang X, Zhang M, Wang Q, Chen X, Yu R, Bao H, Liu J, Wu X, Shao Y, Liang B, Lu K. Real-world circulating tumor DNA analysis depicts resistance mechanism and clonal evolution in ALK inhibitor-treated lung adenocarcinoma patients. ESMO Open 2022; 7(1): 100337

[96]

Rotow J, Bivona TG. Understanding and targeting resistance mechanisms in NSCLC. Nat Rev Cancer 2017; 17(11): 637–658

[97]

Zandi R, Larsen AB, Andersen P, Stockhausen MT, Poulsen HS. Mechanisms for oncogenic activation of the epidermal growth factor receptor. Cell Signal 2007; 19(10): 2013–2023

[98]

Tricker EM, Xu C, Uddin S, Capelletti M, Ercan D, Ogino A, Pratilas CA, Rosen N, Gray NS, Wong KK, Jänne PA. Combined EGFR/MEK inhibition prevents the emergence of resistance in EGFR-mutant lung cancer. Cancer Discov 2015; 5(9): 960–971

[99]

Ho CC, Liao WY, Lin CA, Shih JY, Yu CJ, Yang JC. Acquired BRAF V600E mutation as resistant mechanism after treatment with osimertinib. J Thorac Oncol 2017; 12(3): 567–572

[100]

Gold KA, Lee JJ, Harun N, Tang X, Price J, Kawedia JD, Tran HT, Erasmus JJ, Blumenschein GR, William WN, Wistuba II, Johnson FM. A phase I/II study combining erlotinib and dasatinib for non-small cell lung cancer. Oncologist 2014; 19(10): 1040–1041

[101]

Zhang Z, Lee JC, Lin L, Olivas V, Au V, LaFramboise T, Abdel-Rahman M, Wang X, Levine AD, Rho JK, Choi YJ, Choi CM, Kim SW, Jang SJ, Park YS, Kim WS, Lee DH, Lee JS, Miller VA, Arcila M, Ladanyi M, Moonsamy P, Sawyers C, Boggon TJ, Ma PC, Costa C, Taron M, Rosell R, Halmos B, Bivona TG. Activation of the AXL kinase causes resistance to EGFR-targeted therapy in lung cancer. Nat Genet 2012; 44(8): 852–860

[102]

Oh DY, Bang YJ. HER2-targeted therapies—a role beyond breast cancer. Nat Rev Clin Oncol 2020; 17(1): 33–48

[103]

Zhao Y, Murciano-Goroff YR, Xue JY, Ang A, Lucas J, Mai TT, Da Cruz Paula AF, Saiki AY, Mohn D, Achanta P, Sisk AE, Arora KS, Roy RS, Kim D, Li C, Lim LP, Li M, Bahr A, Loomis BR, de Stanchina E, Reis-Filho JS, Weigelt B, Berger M, Riely G, Arbour KC, Lipford JR, Li BT, Lito P. Diverse alterations associated with resistance to KRAS(G12C) inhibition. Nature 2021; 599(7886): 679–683

[104]

Ho CSL, Tüns AI, Schildhaus HU, Wiesweg M, Grüner BM, Hegedus B, Schuler M, Schramm A, Oeck S. HER2 mediates clinical resistance to the KRASG12C inhibitor sotorasib, which is overcome by co-targeting SHP2. Eur J Cancer 2021; 159: 16–23

[105]

Suzuki S, Yonesaka K, Teramura T, Takehara T, Kato R, Sakai H, Haratani K, Tanizaki J, Kawakami H, Hayashi H, Sakai K, Nishio K, Nakagawa K. KRAS inhibitor resistance in MET-amplified KRASG12C non-small cell lung cancer induced by RAS- and non-RAS-mediated cell signaling mechanisms. Clin Cancer Res 2021; 27(20): 5697–5707

[106]

Wang C, Zhang Z, Sun Y, Wang S, Wu M, Ou Q, Xu Y, Chen Z, Shao Y, Liu H, Hou P. 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(1): 390

[107]

Gower A, Hsu WH, Hsu ST, Wang Y, Giaccone G. EMT is associated with, but does not drive resistance to ALK inhibitors among EML4-ALK non-small cell lung cancer. Mol Oncol 2016; 10(4): 601–609

[108]

Offin M, Chan JM, Tenet M, Rizvi HA, Shen R, Riely GJ, Rekhtman N, Daneshbod Y, Quintanal-Villalonga A, Penson A, Hellmann MD, Arcila ME, Ladanyi M, Pe'er D, Kris MG, Rudin CM, Yu HA. Concurrent RB1 and TP53 alterations define a subset of EGFR-mutant lung cancers at risk for histologic transformation and inferior clinical outcomes. J Thorac Oncol 2019; 14(10): 1784–1793

[109]

Marcoux N, Gettinger SN, O’Kane G, Arbour KC, Neal JW, Husain H, Evans TL, Brahmer JR, Muzikansky A, Bonomi PD, Del Prete S, Wurtz A, Farago AF, Dias-Santagata D, Mino-Kenudson M, Reckamp KL, Yu HA, Wakelee HA, Shepherd FA, Piotrowska Z, Sequist LV. EGFR-mutant adenocarcinomas that transform to small-cell lung cancer and other neuroendocrine carcinomas: clinical outcomes. J Clin Oncol 2019; 37(4): 278–285

[110]

Adachi Y, Ito K, Hayashi Y, Kimura R, Tan TZ, Yamaguchi R, Ebi H. Epithelial-to-mesenchymal transition is a cause of both intrinsic and acquired resistance to KRAS G12C inhibitor in KRAS G12C-mutant non-small cell lung cancer. Clin Cancer Res 2020; 26(22): 5962–5973

[111]

Tulchinsky E, Demidov O, Kriajevska M, Barlev NA, Imyanitov E. EMT: a mechanism for escape from EGFR-targeted therapy in lung cancer. Biochim Biophys Acta Rev Cancer 2019; 1871(1): 29–39

[112]

Soothill JF. UK medical research. Lancet 1988; 332(8593): 1054

[113]

Huang X. The potential role of HGF-MET signaling and autophagy in the war of alectinib versus crizotinib against ALK-positive NSCLC. J Exp Clin Cancer Res 2018; 37(1): 33

[114]

Le X, Nilsson M, Goldman J, Reck M, Nakagawa K, Kato T, Ares LP, Frimodt-Moller B, Wolff K, Visseren-Grul C, Heymach JV, Garon EB. Dual EGFR-VEGF pathway inhibition: a promising strategy for patients with EGFR-mutant NSCLC. J Thorac Oncol 2021; 16(2): 205–215

[115]

Itatani Y, Kawada K, Yamamoto T, Sakai Y. Resistance to anti-angiogenic therapy in cancer-alterations to anti-VEGF pathway. Int J Mol Sci 2018; 19(4): 1232

[116]

Zhong J, Li ZX, Zhao J, Duan JC, Bai H, An TT, Yang XD, Wang J. Analysis of BIM (BCL-2 like 11 gene) deletion polymorphism in Chinese non-small cell lung cancer patients. Thorac Cancer 2014; 5(6): 509–516

[117]

Bivona TG, Hieronymus H, Parker J, Chang K, Taron M, Rosell R, Moonsamy P, Dahlman K, Miller VA, Costa C, Hannon G, Sawyers CL. FAS and NF-κB signalling modulate dependence of lung cancers on mutant EGFR. Nature 2011; 471(7339): 523–526

[118]

Budha NR, Frymoyer A, Smelick GS, Jin JY, Yago MR, Dresser MJ, Holden SN, Benet LZ, Ware JA. Drug absorption interactions between oral targeted anticancer agents and PPIs: is pH-dependent solubility the Achilles heel of targeted therapy?. Clin Pharmacol Ther 2012; 92(2): 203–213

[119]

Lim ZF, Ma PC. Emerging insights of tumor heterogeneity and drug resistance mechanisms in lung cancer targeted therapy. J Hematol Oncol 2019; 12(1): 134

[120]

Luo X, Gong X, Su L, Lin H, Yang Z, Yan X, Gao J. Activatable mitochondria-targeting organoarsenic prodrugs for bioenergetic cancer therapy. Angew Chem Int Ed Engl 2021; 60(3): 1403–1410

[121]

Cross DA, Ashton SE, Ghiorghiu S, Eberlein C, Nebhan CA, Spitzler PJ, Orme JP, Finlay MR, Ward RA, Mellor MJ, Hughes G, Rahi A, Jacobs VN, Red Brewer M, Ichihara E, Sun J, Jin H, Ballard P, Al-Kadhimi K, Rowlinson R, Klinowska T, Richmond GH, Cantarini M, Kim DW, Ranson MR, Pao W. AZD9291, an irreversible EGFR TKI, overcomes T790M-mediated resistance to EGFR inhibitors in lung cancer. Cancer Discov 2014; 4(9): 1046–1061

[122]

Giroux-Leprieur E, Dumenil C, Chinet T. Combination of crizotinib and osimertinib or erlotinib might overcome MET-mediated resistance to EGFR tyrosine kinase inhibitor in EGFR-mutated adenocarcinoma. J Thorac Oncol 2018; 13(11): e232–e234

[123]

Piotrowska Z, Isozaki H, Lennerz JK, Gainor JF, Lennes IT, Zhu VW, Marcoux N, Banwait MK, Digumarthy SR, Su W, Yoda S, Riley AK, Nangia V, Lin JJ, Nagy RJ, Lanman RB, Dias-Santagata D, Mino-Kenudson M, Iafrate AJ, Heist RS, Shaw AT, Evans EK, Clifford C, Ou SI, Wolf B, Hata AN, Sequist LV. 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(12): 1529–1539

[124]

Wu SG, Shih JY. Management of acquired resistance to EGFR TKI-targeted therapy in advanced non-small cell lung cancer. Mol Cancer 2018; 17(1): 38

[125]

Yu L, Bazhenova L, Gold K, Tran L, Hilburn V, Vu P, Patel SP. Clinicopathologic and molecular characteristics of EGFR-mutant lung adenocarcinomas that transform to small cell lung cancer after TKI therapy. Transl Lung Cancer Res 2022; 11(3): 452–461

[126]

Arulananda S, Do H, Musafer A, Mitchell P, Dobrovic A, John T. Combination osimertinib and gefitinib in C797S and T790M EGFR-mutated non-small cell lung cancer. J Thorac Oncol 2017; 12(11): 1728–1732

[127]

Zhao J, Zou M, Lv J, Han Y, Wang G, Wang G. Effective treatment of pulmonary adenocarcinoma harboring triple EGFR mutations of L858R, T790M, and cis-C797S by osimertinib, bevacizumab, and brigatinib combination therapy: a case report. OncoTargets Ther 2018; 11: 5545–5550

[128]

Scalvini L, Castelli R, La Monica S, Tiseo M, Alfieri R. Fighting tertiary mutations in EGFR-driven lung-cancers: current advances and future perspectives in medicinal chemistry. Biochem Pharmacol 2021; 190: 114643

[129]

Neijssen J, Cardoso RMF, Chevalier KM, Wiegman L, Valerius T, Anderson GM, Moores SL, Schuurman J, Parren PWHI, Strohl WR, Chiu ML. Discovery of amivantamab (JNJ-61186372), a bispecific antibody targeting EGFR and MET. J Biol Chem 2021; 296: 100641

[130]

Sato H, Yamamoto H, Sakaguchi M, Shien K, Tomida S, Shien T, Ikeda H, Hatono M, Torigoe H, Namba K, Yoshioka T, Kurihara E, Ogoshi Y, Takahashi Y, Soh J, Toyooka S. Combined inhibition of MEK and PI3K pathways overcomes acquired resistance to EGFR-TKIs in non-small cell lung cancer. Cancer Sci 2018; 109(10): 3183–3196

[131]

Zhou C, Li X, Wang Q, Gao G, Zhang Y, Chen J, Shu Y, Hu Y, Fan Y, Fang J, Chen G, Zhao J, He J, Wu F, Zou J, Zhu X, Lin X. Pyrotinib in HER2-mutant advanced lung adenocarcinoma after platinum-based chemotherapy: a multicenter, open-label, single-arm, phase II study. J Clin Oncol 2020; 38(24): 2753–2761

[132]

Watanabe S, Yoshida T, Kawakami H, Takegawa N, Tanizaki J, Hayashi H, Takeda M, Yonesaka K, Tsurutani J, Nakagawa K. T790M-selective EGFR-TKI combined with dasatinib as an optimal strategy for overcoming EGFR-TKI resistance in T790M-positive non-small cell lung cancer. Mol Cancer Ther 2017; 16(11): 2563–2571

[133]

Okura N, Nishioka N, Yamada T, Taniguchi H, Tanimura K, Katayama Y, Yoshimura A, Watanabe S, Kikuchi T, Shiotsu S, Kitazaki T, Nishiyama A, Iwasaku M, Kaneko Y, Uchino J, Uehara H, Horinaka M, Sakai T, Tanaka K, Kozaki R, Yano S, Takayama K. ONO-7475, a novel AXL inhibitor, suppresses the adaptive resistance to initial EGFR-TKI treatment in EGFR-mutated non-small cell lung cancer. Clin Cancer Res 2020; 26(9): 2244–2256

[134]

Ramalingam SS, Cheng Y, Zhou C, Ohe Y, Imamura F, Cho BC, Lin MC, Majem M, Shah R, Rukazenkov Y, Todd A, Markovets A, Barrett JC, Chmielecki J, Gray J. Mechanisms of acquired resistance to first-line osimertinib: preliminary data from the phase III FLAURA study. Ann Oncol 2018; 29(suppl_8): VIII740

[135]

Yin X, Li Y, Wang H, Jia T, Wang E, Luo Y, Wei Y, Qin Z, Ma X. Small cell lung cancer transformation: from pathogenesis to treatment. Semin Cancer Biol 2022; 86(Pt 2): 595–606

[136]

Schoenfeld AJ, Chan JM, Kubota D, Sato H, Rizvi H, Daneshbod Y, Chang JC, Paik PK, Offin M, Arcila ME, Davare MA, Shinde U, Pe′er D, Rekhtman N, Kris MG, Somwar R, Riely GJ, Ladanyi M, Yu HA. Tumor analyses reveal squamous transformation and off-target alterations as early resistance mechanisms to first-line osimertinib in EGFR-mutant lung cancer. Clin Cancer Res 2020; 26(11): 2654–2663

[137]

Russo A, Cardona AF, Caglevic C, Manca P, Ruiz-Patiño A, Arrieta O, Rolfo C. Overcoming TKI resistance in fusion-driven NSCLC: new generation inhibitors and rationale for combination strategies. Transl Lung Cancer Res 2020; 9(6): 2581–2598

[138]

Shaw AT, Solomon BJ, Besse B, Bauer TM, Lin CC, Soo RA, Riely GJ, Ou SI, Clancy JS, Li S, Abbattista A, Thurm H, Satouchi M, Camidge DR, Kao S, Chiari R, Gadgeel SM, Felip E, Martini JF. ALK resistance mutations and efficacy of lorlatinib in advanced anaplastic lymphoma kinase-positive non-small-cell lung cancer. J Clin Oncol 2019; 37(16): 1370–1379

[139]

Murray BW, Zhai D, Deng W, Zhang X, Ung J, Nguyen V, Zhang H, Barrera M, Parra A, Cowell J, Lee DJ, Aloysius H, Rogers E. TPX-0131, a potent CNS-penetrant, next-generation inhibitor of wild-type ALK and ALK-resistant mutations. Mol Cancer Ther 2021; 20(9): 1499–1507

[140]

Dagogo-Jack I, Yoda S, Lennerz JK, Langenbucher A, Lin JJ, Rooney MM, Prutisto-Chang K, Oh A, Adams NA, Yeap BY, Chin E, Do A, Marble HD, Stevens SE, Digumarthy SR, Saxena A, Nagy RJ, Benes CH, Azzoli CG, Lawrence MS, Gainor JF, Shaw AT, Hata AN. MET alterations are a recurring and actionable resistance mechanism in ALK-positive lung cancer. Clin Cancer Res 2020; 26(11): 2535–2545

[141]

McCoach CE, Le AT, Gowan K, Jones K, Schubert L, Doak A, Estrada-Bernal A, Davies KD, Merrick DT, Bunn PA Jr, Purcell WT, Dziadziuszko R, Varella-Garcia M, Aisner DL, Camidge DR, Doebele RC. Resistance mechanisms to targeted therapies in ROS1+ and ALK+ non-small cell lung cancer. Clin Cancer Res 2018; 24(14): 3334–3347

[142]

Facchinetti F, Loriot Y, Kuo MS, Mahjoubi L, Lacroix L, Planchard D, Besse B, Farace F, Auger N, Remon J, Scoazec JY, André F, Soria JC, Friboulet L. Crizotinib-resistant ROS1 mutations reveal a predictive kinase inhibitor sensitivity model for ROS1- and ALK-rearranged lung cancers. Clin Cancer Res 2016; 22(24): 5983–5991

[143]

Katayama R, Shaw AT, Khan TM, Mino-Kenudson M, Solomon BJ, Halmos B, Jessop NA, Wain JC, Yeo AT, Benes C, Drew L, Saeh JC, Crosby K, Sequist LV, Iafrate AJ, Engelman JA. Mechanisms of acquired crizotinib resistance in ALK-rearranged lung Cancers. Sci Transl Med 2012; 4(120): 120ra17

[144]

Drilon A, Somwar R, Wagner JP, Vellore NA, Eide CA, Zabriskie MS, Arcila ME, Hechtman JF, Wang L, Smith RS, Kris MG, Riely GJ, Druker BJ, O’Hare T, Ladanyi M, Davare MA. A novel crizotinib-resistant solvent-front mutation responsive to cabozantinib therapy in a patient with ROS1-rearranged lung cancer. Clin Cancer Res 2016; 22(10): 2351–2358

[145]

Gainor JF, Tseng D, Yoda S, Dagogo-Jack I, Friboulet L, Lin JJ, Hubbeling HG, Dardaei L, Farago AF, Schultz KR, Ferris LA, Piotrowska Z, Hardwick J, Huang D, Mino-Kenudson M, Iafrate AJ, Hata AN, Yeap BY, Shaw AT. Patterns of metastatic spread and mechanisms of resistance to crizotinib in ROS1-positive non-small-cell lung cancer. JCO Precis Oncol 2017; 2017: PO.17.00063

[146]

Cui M, Han Y, Li P, Zhang J, Ou Q, Tong X, Zhao R, Dong N, Wu X, Li W, Jiang G. Molecular and clinicopathological characteristics of ROS1-rearranged non-small-cell lung cancers identified by next-generation sequencing. Mol Oncol 2020; 14(11): 2787–2795

[147]

Subbiah V, Shen T, Terzyan SS, Liu X, Hu X, Patel KP, Hu M, Cabanillas M, Behrang A, Meric-Bernstam F, Vo PTT, Mooers BHM, Wu J. Structural basis of acquired resistance to selpercatinib and pralsetinib mediated by non-gatekeeper RET mutations. Ann Oncol 2021; 32(2): 261–268

[148]

Nelson-Taylor SK, Le AT, Yoo M, Schubert L, Mishall KM, Doak A, Varella-Garcia M, Tan AC, Doebele RC. Resistance to RET-inhibition in RET-rearranged NSCLC is mediated by reactivation of RAS/MAPK signaling. Mol Cancer Ther 2017; 16(8): 1623–1633

[149]

Drusbosky LM, Rodriguez E, Dawar R, Ikpeazu CV. Therapeutic strategies in RET gene rearranged non-small cell lung cancer. J Hematol Oncol 2021; 14(1): 50

[150]

Shimizu Y, Maruyama K, Suzuki M, Kawachi H, Low SK, Oh-Hara T, Takeuchi K, Fujita N, Nagayama S, Katayama R. Acquired resistance to BRAF inhibitors is mediated by BRAF splicing variants in BRAF V600E mutation-positive colorectal neuroendocrine carcinoma. Cancer Lett 2022; 543: 215799

[151]

Kulkarni A, Al-Hraishawi H, Simhadri S, Hirshfield KM, Chen S, Pine S, Jeyamohan C, Sokol L, Ali S, Teo ML, White E, Rodriguez-Rodriguez L, Mehnert JM, Ganesan S. BRAF fusion as a novel mechanism of acquired resistance to vemurafenib in BRAFV600E mutant melanoma. Clin Cancer Res 2017; 23(18): 5631–5638

[152]

Johnson DB, Menzies AM, Zimmer L, Eroglu Z, Ye F, Zhao S, Rizos H, Sucker A, Scolyer RA, Gutzmer R, Gogas H, Kefford RF, Thompson JF, Becker JC, Berking C, Egberts F, Loquai C, Goldinger SM, Pupo GM, Hugo W, Kong X, Garraway LA, Sosman JA, Ribas A, Lo RS, Long GV, Schadendorf D. Acquired BRAF inhibitor resistance: a multicenter meta-analysis of the spectrum and frequencies, clinical behaviour, and phenotypic associations of resistance mechanisms. Eur J Cancer 2015; 51(18): 2792–2799

[153]

Proietti I, Skroza N, Bernardini N, Tolino E, Balduzzi V, Marchesiello A, Michelini S, Volpe S, Mambrin A, Mangino G, Romeo G, Maddalena P, Rees C, Potenza C. Mechanisms of acquired BRAF inhibitor resistance in melanoma: a systematic review. Cancers (Basel) 2020; 12(10): 2801

[154]

Rudin CM, Hong K, Streit M. Molecular characterization of acquired resistance to the BRAF inhibitor dabrafenib in a patient with BRAF-mutant non-small-cell lung cancer. J Thorac Oncol 2013; 8(5): e41–e42

[155]

Ding G, Wang J, Ding P, Wen Y, Yang L. Case report: HER2 amplification as a resistance mechanism to crizotinib in NSCLC with MET exon 14 skipping. Cancer Biol Ther 2019; 20(6): 837–842

[156]

Bahcall M, Awad MM, Sholl LM, Wilson FH, Xu M, Wang S, Palakurthi S, Choi J, Ivanova EV, Leonardi GC, Ulrich BC, Paweletz CP, Kirschmeier PT, Watanabe M, Baba H, Nishino M, Nagy RJ, Lanman RB, Capelletti M, Chambers ES, Redig AJ, VanderLaan PA, Costa DB, Imamura Y, Jänne PA. Amplification of wild-type KRAS imparts resistance to crizotinib in MET exon 14 mutant non-small cell lung cancer. Clin Cancer Res 2018; 24(23): 5963–5976

[157]

Dagogo-Jack I, Fabrizio D, Lennerz J, Schrock AB, Young L, Mino-Kenudson M, Digumarthy SR, Heist RS, Ali SM, Miller VA, Shaw AT. Circulating tumor DNA identifies EGFR coamplification as a mechanism of resistance to crizotinib in a patient with advanced MET-amplified lung adenocarcinoma. J Thorac Oncol 2017; 12(10): e155–e157

[158]

Yu Y, Ren Y, Fang J, Cao L, Liang Z, Guo Q, Han S, Ji Z, Wang Y, Sun Y, Chen Y, Li X, Xu H, Zhou J, Jiang L, Cheng Y, Han Z, Shi J, Chen G, Ma R, Fan Y, Sun S, Jiao L, Jia X, Wang L, Lu P, Li J, Xu Q, Luo X, Su W, Lu S. ctDNA analysis in the savolitinib phase II study in non-small cell lung cancer (NSCLC) patients (pts) harboring MET exon 14 skipping alterations (METex14). Cancer Res 2021; 81(13_Supplement): CT158

[159]

Shen B, Wu F, Ye J, Liang R, Wang R, Yu R, Wu X, Shao YW, Feng J. Crizotinib-resistant MET mutations in gastric cancer patients are sensitive to type II tyrosine kinase inhibitors. Future Oncol 2019; 15(22): 2585–2593

[160]

Guo R, Offin M, Brannon AR, Chang J, Chow A, Delasos L, Girshman J, Wilkins O, McCarthy CG, Makhnin A, Falcon C, Scott K, Tian Y, Cecchi F, Hembrough T, Alex D, Shen R, Benayed R, Li BT, Rudin CM, Kris MG, Arcila ME, Rekhtman N, Paik P, Zehir A, Drilon A. MET exon 14-altered lung cancers and MET inhibitor resistance. Clin Cancer Res 2021; 27(3): 799–806

[161]

Koga T, Kobayashi Y, Tomizawa K, Suda K, Kosaka T, Sesumi Y, Fujino T, Nishino M, Ohara S, Chiba M, Shimoji M, Takemoto T, Suzuki M, Jänne PA, Mitsudomi T. Activity of a novel HER2 inhibitor, poziotinib, for HER2 exon 20 mutations in lung cancer and mechanism of acquired resistance: an in vitro study. Lung Cancer 2018; 126: 72–79

[162]

Chuang JC, Stehr H, Liang Y, Das M, Huang J, Diehn M, Wakelee HA, Neal JW. ERBB2-mutated metastatic non-small cell lung cancer: response and resistance to targeted therapies. J Thorac Oncol 2017; 12(5): 833–842

[163]

Yu X, Wang T, Lou Y, Li Y. Combination of in silico analysis and in vitro assay to investigate drug response to human epidermal growth factor receptor 2 mutations in lung cancer. Mol Inform 2016; 35(1): 25–35

[164]

Antonia SJ, Villegas A, Daniel D, Vicente D, Murakami S, Hui R, Yokoi T, Chiappori A, Lee KH, de Wit M, Cho BC, Bourhaba M, Quantin X, Tokito T, Mekhail T, Planchard D, Kim YC, Karapetis CS, Hiret S, Ostoros G, Kubota K, Gray JE, Paz-Ares L, de Castro Carpeño J, Wadsworth C, Melillo G, Jiang H, Huang Y, Dennis PA, Özgüroğlu M; PACIFIC Investigators. Durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. N Engl J Med 2017; 377(20): 1919–1929

[165]

Antonia SJ, Villegas A, Daniel D, Vicente D, Murakami S, Hui R, Kurata T, Chiappori A, Lee KH, de Wit M, Cho BC, Bourhaba M, Quantin X, Tokito T, Mekhail T, Planchard D, Kim YC, Karapetis CS, Hiret S, Ostoros G, Kubota K, Gray JE, Paz-Ares L, de Castro Carpeño J, Faivre-Finn C, Reck M, Vansteenkiste J, Spigel DR, Wadsworth C, Melillo G, Taboada M, Dennis PA, Özgüroğlu M; PACIFIC Investigators. Overall survival with durvalumab after chemoradiotherapy in stage III NSCLC. N Engl J Med 2018; 379(24): 2342–2350

[166]

Faivre-Finn C, Vicente D, Kurata T, Planchard D, Paz-Ares L, Vansteenkiste JF, Spigel DR, Garassino MC, Reck M, Senan S, Naidoo J, Rimner A, Wu YL, Gray JE, Özgüroğlu M, Lee KH, Cho BC, Kato T, de Wit M, Newton M, Wang L, Thiyagarajah P, Antonia SJ. Four-year survival with durvalumab after chemoradiotherapy in stage III NSCLC—an update from the PACIFIC trial. J Thorac Oncol 2021; 16(5): 860–867

[167]

Lee CK, Man J, Lord S, Cooper W, Links M, Gebski V, Herbst RS, Gralla RJ, Mok T, Yang JC. Clinical and molecular characteristics associated with survival among patients treated with checkpoint inhibitors for advanced non-small cell lung carcinoma: a systematic review and meta-analysis. JAMA Oncol 2018; 4(2): 210–216

[168]

Garassino MC, Cho BC, Kim JH, Mazières J, Vansteenkiste J, Lena H, Corral Jaime J, Gray JE, Powderly J, Chouaid C, Bidoli P, Wheatley-Price P, Park K, Soo RA, Huang Y, Wadsworth C, Dennis PA, Rizvi NA; ATLANTIC Investigators. 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(4): 521–536

[169]

Hayashi H, Chiba Y, Sakai K, Fujita T, Yoshioka H, Sakai D, Kitagawa C, Naito T, Takeda K, Okamoto I, Mitsudomi T, Kawakami Y, Nishio K, Nakamura S, Yamamoto N, Nakagawa K. A randomized phase II study comparing nivolumab with carboplatin-pemetrexed for patients with EGFR mutation-positive nonsquamous non-small-cell lung cancer who acquire resistance to tyrosine kinase inhibitors not due to a secondary T790M mutation: rationale and protocol design for the WJOG8515L study. Clin Lung Cancer 2017; 18(6): 719–723

[170]

Streicher K, Morehouse C, Sebastian Y, Kuziora M, Higgs BW, Ranade K. Gene expression analysis of tumor biopsies from a trial of durvalumab to identify subsets of NSCLC with shared immune pathways. J Clin Oncol 2017; 35(15 suppl): 3041

[171]

Martin P, Spitzmueller A, Wu S, Widmaier M, Korn R, Althammer S, Zha J, Higgs BW, Cooper Z, Steele K. Mutually exclusive expression of CD73 and PDL1 in tumors from patients (pt) with NSCLC, gastroesophageal (GE) and urothelial bladder carcinoma (UBC). J Clin Oncol 2017; 35(15 suppl): 3079

[172]

Offin M, Rizvi H, Tenet M, Ni A, Sanchez-Vega F, Li BT, Drilon A, Kris MG, Rudin CM, Schultz N, Arcila ME, Ladanyi M, Riely GJ, Yu H, Hellmann MD. Tumor mutation burden and efficacy of EGFR-tyrosine kinase inhibitors in patients with EGFR-mutant lung cancers. Clin Cancer Res 2019; 25(3): 1063–1069

[173]

Haratani K, Hayashi H, Tanaka T, Kaneda H, Togashi Y, Sakai K, Hayashi K, Tomida S, Chiba Y, Yonesaka K, Nonagase Y, Takahama T, Tanizaki J, Tanaka K, Yoshida T, Tanimura K, Takeda M, Yoshioka H, Ishida T, Mitsudomi T, Nishio K, Nakagawa K. Tumor immune microenvironment and nivolumab efficacy in EGFR mutation-positive non-small-cell lung cancer based on T790M status after disease progression during EGFR-TKI treatment. Ann Oncol 2017; 28(7): 1532–1539

[174]

Isomoto K, Haratani K, Hayashi H, Shimizu S, Tomida S, Niwa T, Yokoyama T, Fukuda Y, Chiba Y, Kato R, Tanizaki J, Tanaka K, Takeda M, Ogura T, Ishida T, Ito A, Nakagawa K. Impact of EGFR-TKI treatment on the tumor immune microenvironment in EGFR mutation-positive non-small cell lung cancer. Clin Cancer Res 2020; 26(8): 2037–2046

[175]

Liu S, Wu F, Li X, Zhao C, Jia Y, Jia K, Han R, Qiao M, Li W, Yu J, Zhou F, Xiong A, Chen B, Fan J, Ren S, Zhou C. 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

[176]

Hastings K, Yu HA, Wei W, Sanchez-Vega F, DeVeaux M, Choi J, Rizvi H, Lisberg A, Truini A, Lydon CA, Liu Z, Henick BS, Wurtz A, Cai G, Plodkowski AJ, Long NM, Halpenny DF, Killam J, Oliva I, Schultz N, Riely GJ, Arcila ME, Ladanyi M, Zelterman D, Herbst RS, Goldberg SB, Awad MM, Garon EB, Gettinger S, Hellmann MD, Politi K. EGFR mutation subtypes and response to immune checkpoint blockade treatment in non-small-cell lung cancer. Ann Oncol 2019; 30(8): 1311–1320

[177]

Seegobin K, Majeed U, Wiest N, Manochakian R, Lou Y, Zhao Y. Immunotherapy in non-small cell lung cancer with actionable mutations other than EGFR. Front Oncol 2021; 11: 750657

[178]

Mazieres J, Drilon A, Lusque A, Mhanna L, Cortot AB, Mezquita L, Thai AA, Mascaux C, Couraud S, Veillon R, Van den Heuvel M, Neal J, Peled N, Früh M, Ng TL, Gounant V, Popat S, Diebold J, Sabari J, Zhu VW, Rothschild SI, Bironzo P, Martinez-Marti A, Curioni-Fontecedro A, Rosell R, Lattuca-Truc M, Wiesweg M, Besse B, Solomon B, Barlesi F, Schouten RD, Wakelee H, Camidge DR, Zalcman G, Novello S, Ou SI, Milia J, Gautschi O. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: results from the IMMUNOTARGET registry. Ann Oncol 2019; 30(8): 1321–1328

[179]

Tanaka I, Morise M. Current immunotherapeutic strategies targeting the PD-1/PD-L1 axis in non-small cell lung cancer with oncogenic driver mutations. Int J Mol Sci 2021; 23(1): 245

[180]

Negrao MV, Skoulidis F, Montesion M, Schulze K, Bara I, Shen V, Xu H, Hu S, Sui D, Elamin YY, Le X, Goldberg ME, Murugesan K, Wu CJ, Zhang J, Barreto DS, Robichaux JP, Reuben A, Cascone T, Gay CM, Mitchell KG, Hong L, Rinsurongkawong W, Roth JA, Swisher SG, Lee J, Tsao A, Papadimitrakopoulou V, Gibbons DL, Glisson BS, Singal G, Miller VA, Alexander B, Frampton G, Albacker LA, Shames D, Zhang J, Heymach JV. Oncogene-specific differences in tumor mutational burden, PD-L1 expression, and outcomes from immunotherapy in non-small cell lung cancer. J Immunother Cancer 2021; 9(8): e002891

[181]

Ricciuti B, Son J, Okoro JJ, Mira A, Patrucco E, Eum Y, Wang X, Paranal R, Wang H, Lin M, Haikala HM, Li J, Xu Y, Alessi JV, Chhoeu C, Redig AJ, Köhler J, Dholakia KH, Chen Y, Richard E, Nokin MJ, Santamaria D, Gokhale PC, Awad MM, Jänne PA, Ambrogio C. Comparative analysis and isoform-specific therapeutic vulnerabilities of KRAS Mutations in non-small cell lung cancer. Clin Cancer Res 2022; 28(8): 1640–1650

[182]

Kumagai S, Koyama S, Nishikawa H. Antitumour immunity regulated by aberrant ERBB family signalling. Nat Rev Cancer 2021; 21(3): 181–197

[183]

Gavralidis A, Gainor JF. Immunotherapy in EGFR-mutant and ALK-positive lung cancer: implications for oncogene-driven lung cancer. Cancer J 2020; 26(6): 517–524

[184]

To KKW, Fong W, Cho WCS. Immunotherapy in treating EGFR-mutant lung cancer: current challenges and new strategies. Front Oncol 2021; 11: 635007

[185]

Yang JC, Shepherd FA, Kim DW, Lee GW, Lee JS, Chang GC, Lee SS, Wei YF, Lee YG, Laus G, Collins B, Pisetzky F, Horn L. Osimertinib plus durvalumab versus osimertinib monotherapy in EGFR T790M-positive NSCLC following previous EGFR TKI therapy: CAURAL brief report. J Thorac Oncol 2019; 14(5): 933–939

[186]

Gettinger S, Hellmann MD, Chow LQM, Borghaei H, Antonia S, Brahmer JR, Goldman JW, Gerber DE, Juergens RA, Shepherd FA, Laurie SA, Young TC, Li X, Geese WJ, Rizvi N. Nivolumab plus erlotinib in patients with EGFR-mutant advanced NSCLC. J Thorac Oncol 2018; 13(9): 1363–1372

[187]

Oxnard GR, Yang JC, Yu H, Kim SW, Saka H, Horn L, Goto K, Ohe Y, Mann H, Thress KS, Frigault MM, Vishwanathan K, Ghiorghiu D, Ramalingam SS, Ahn MJ. TATTON: a multi-arm, phase Ib trial of osimertinib combined with selumetinib, savolitinib, or durvalumab in EGFR-mutant lung cancer. Ann Oncol 2020; 31(4): 507–516

[188]

Creelan BC, Yeh TC, Kim SW, Nogami N, Kim DW, Chow LQM, Kanda S, Taylor R, Tang W, Tang M, Angell HK, Roudier MP, Marotti M, Gibbons DL. A Phase 1 study of gefitinib combined with durvalumab in EGFR TKI-naive patients with EGFR mutation-positive locally advanced/metastatic non-small-cell lung cancer. Br J Cancer 2021; 124(2): 383–390

[189]

Yang JC, Gadgeel SM, Sequist LV, Wu CL, Papadimitrakopoulou VA, Su WC, Fiore J, Saraf S, Raftopoulos H, Patnaik A. Pembrolizumab in combination with erlotinib or gefitinib as first-line therapy for advanced NSCLC with sensitizing EGFR mutation. J Thorac Oncol 2019; 14(3): 553–559

[190]

AntoniaSJRivzi NAChowLQBorghaeiHBrahmerJR JuergensRShepherd FAShepherdSAGerberDEGerberJ ShenYHarbison CHarbisonACGettingerS. Nivolumab (anti-PD-1; BMS-936558, ONO-4538) in combination with platinumbased doublet chemotherapy (Pt-DC) or erlotinib in advanced non small cell lung cancer (NSCLC). J Thorac Oncol 2014; 32(5s): abstr 8113

[191]

Cocco E, Scaltriti M, Drilon A. NTRK fusion-positive cancers and TRK inhibitor therapy. Nat Rev Clin Oncol 2018; 15(12): 731–747

[192]

Schoenfeld AJ, Arbour KC, Rizvi H, Iqbal AN, Gadgeel SM, Girshman J, Kris MG, Riely GJ, Yu HA, Hellmann MD. Severe immune-related adverse events are common with sequential PD-(L)1 blockade and osimertinib. Ann Oncol 2019; 30(5): 839–844

[193]

Latif H, Liu SV. Combining immunotherapy and epidermal growth factor receptor kinase inhibitors: worth the risk?. Ann Transl Med 2019; 7(Suppl 3): S76

[194]

Reck M, Mok TSK, Nishio M, Jotte RM, Cappuzzo F, Orlandi F, Stroyakovskiy D, Nogami N, Rodríguez-Abreu D, Moro-Sibilot D, Thomas CA, Barlesi F, Finley G, Lee A, Coleman S, Deng Y, Kowanetz M, Shankar G, Lin W, Socinski MA, Reck M, Mok TSK, Nishio M, Jotte RM, Cappuzzo F, Orlandi F, Stroyakovskiy D, Nogami N, Rodríguez-Abreu D, Moro-Sibilot D, Thomas CA, Barlesi F, Finley G, Lee A, Coleman S, Deng Y, Kowanetz M, Shankar G, Lin W, Socinski MA; IMpower150 Study Group. 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(5): 387–401

[195]

Nagasaka M, Ou SI. ORIENT-31 as the Sakigake “Charging Samurai” born of IMpower150 but will MARIPOSA-2 IMPRESS in the “Meiji Modernization” of post-3G EGFR TKI progression?. Lung Cancer (Auckl) 2022; 13: 13–21

[196]

Gadgeel S, Dziubek K, Nagasaka M, Braun T, Hassan K, Cheng H, Wozniak A, Halmos B, Stevenson J, Patil P, Pennell N, Fidler MJ, Bonomi P, Qin A, Niu Z, Nagrath S, Kalemkerian G. OA09.03 Pembrolizumab in combination with platinum-based chemotherapy in recurrent EGFR/ALK-positive non-small cell lung cancer (NSCLC). J Thorac Oncol 2021; 16(10): S863

[197]

Han B, Tian P, Zhao Y, Yu X, Guo Q, Yu Z, Zhang X, Li Y, Chen L, Shi X, Zhang Y, Wang J. 148P A phase II study of tislelizumab plus chemotherapy in EGFR mutated advanced non-squamous NSCLC patients failed to EGFR TKI therapies: first analysis. Ann Oncol 2021; 32: S1443–S1444

[198]

Zhou C, Gao G, Wu F, Chen X, Li W, Xiong A, Su CX, Cai W, Ren S, Jiang T, Wang YN, Kang X, Wang Q. A phase Ib study of SHR-1210 plus apatinib for heavily previously treated advanced non-squamous non-small cell lung cancer (NSCLC) patients. J Clin Oncol 2018; 36(15 suppl): e21017

[199]

Lipson EJ, Tawbi H A-H, Schadendorf D, Ascierto PA, Matamala L, Gutiérrez EC, Rutkowski P, Gogas H, Lao CD, de Menezes JJ, Dalle S, Arance AM, Grob J-J, Srivastava S, Abaskharoun M, Simonsen KL, Li B, Long GV, Hodi FS. Relatlimab (RELA) plus nivolumab (NIVO) versus NIVO in first-line advanced melanoma: primary phase III results from RELATIVITY-047 (CA224-047). J Clin Oncol 2021; 39(15_suppl): 9503

[200]

Harding JJ, Patnaik A, Moreno V, Stein M, Jankowska AM, de Mendizabal NV, Liu ZT, Koneru M, Calvo E. A phase Ia/Ib study of an anti-TIM-3 antibody (LY3321367) monotherapy or in combination with an anti-PD-L1 antibody (LY3300054): interim safety, efficacy, and pharmacokinetic findings in advanced cancers. J Clin Oncol 2019; 37(8_suppl): 12

[201]

Rodriguez-Abreu D, Johnson ML, Hussein MA, Cobo M, Patel AJ, Secen NM, Lee KH, Massuti B, Hiret S, Yang J C-H, Barlesi F, Lee DH, Paz-Ares LG, Hsieh RW, Miller K, Patil N, Twomey P, Kapp AV, Meng R, Cho BC. Primary analysis of a randomized, double-blind, phase II study of the anti-TIGIT antibody tiragolumab (tira) plus atezolizumab (atezo) versus placebo plus atezo as first-line (1L) treatment in patients with PD-L1-selected NSCLC (CITYSCAPE). J Clin Oncol 2020; 38(15_suppl): 9503

[202]

Jin R, Liu L, Xing Y, Meng T, Ma L, Pei J, Cong Y, Zhang X, Ren Z, Wang X, Shen J, Yu K. Dual mechanisms of novel CD73-targeted antibody and antibody-drug conjugate in inhibiting lung tumor growth and promoting antitumor immune-effector function. Mol Cancer Ther 2020; 19(11): 2340–2352

[203]

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

[204]

Genova C, Dellepiane C, Carrega P, Sommariva S, Ferlazzo G, Pronzato P, Gangemi R, Filaci G, Coco S, Croce M. Therapeutic implications of tumor microenvironment in lung cancer: focus on immune checkpoint blockade. Front Immunol 2022; 12: 799455

[205]

Montisci A, Vietri MT, Palmieri V, Sala S, Donatelli F, Napoli C. Cardiac toxicity associated with cancer immunotherapy and biological drugs. Cancers (Basel) 2021; 13(19): 4797

[206]

Xia L, Wen L, Wang S. SHP2 inhibition benefits epidermal growth factor receptor-mutated non-small cell lung cancer therapy. Mini Rev Med Chem 2021; 21(11): 1314–1321

[207]

Song Y, Zhao M, Zhang H, Yu B. Double-edged roles of protein tyrosine phosphatase SHP2 in cancer and its inhibitors in clinical trials. Pharmacol Ther 2022; 230: 107966

[208]

Laskin J, Liu SV, Tolba K, Heining C, Schlenk RF, Cheema P, Cadranel J, Jones MR, Drilon A, Cseh A, Gyorffy S, Solca F, Duruisseaux M. NRG1 fusion-driven tumors: biology, detection, and the therapeutic role of afatinib and other ErbB-targeting agents. Ann Oncol 2020; 31(12): 1693–1703

[209]

Rosas D, Raez LE, Russo A, Rolfo C. Neuregulin 1 gene (NRG1). A potentially new targetable alteration for the treatment of lung cancer. Cancers (Basel) 2021; 13(20): 5038

[210]

Gan HK, Millward M, Jalving M, Garrido-Laguna I, Lickliter JD, Schellens JHM, Lolkema MP, Van Herpen CLM, Hug B, Tang L, O’Connor-Semmes R, Gagnon R, Ellis C, Ganji G, Matheny C, Drilon A. A phase I, first-in-human study of GSK2849330, an anti-HER3 monoclonal antibody, in HER3-expressing solid tumors. Oncologist 2021; 26(10): e1844–e1853

[211]

Jänne PA, Baik C, Su WC, Johnson ML, Hayashi H, Nishio M, Kim DW, Koczywas M, Gold KA, Steuer CE, Murakami H, Yang JC, Kim SW, Vigliotti M, Shi R, Qi Z, Qiu Y, Zhao L, Sternberg D, Yu C, Yu HA. Efficacy and safety of patritumab deruxtecan (HER3-DXd) in EGFR inhibitor-resistant, EGFR-mutated non-small cell lung cancer. Cancer Discov 2022; 12(1): 74–89

RIGHTS & PERMISSIONS

Higher Education Press

AI Summary AI Mindmap
PDF (1728KB)

5588

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/