Next Article in Journal
Two Siblings Homozygous for F508del-CFTR Have Varied Disease Phenotypes and Protein Biomarkers
Next Article in Special Issue
Immune Stroma in Lung Cancer and Idiopathic Pulmonary Fibrosis: A Common Biologic Landscape?
Previous Article in Journal
Effects of a Catechol-Functionalized Hyaluronic Acid Patch Combined with Human Adipose-Derived Stem Cells in Diabetic Wound Healing
Previous Article in Special Issue
The CD44high Subpopulation of Multifraction Irradiation-Surviving NSCLC Cells Exhibits Partial EMT-Program Activation and DNA Damage Response Depending on Their p53 Status
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Novel Emerging Molecular Targets in Non-Small Cell Lung Cancer

1
Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
2
Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, 16132 Genoa, Italy
3
Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
4
Medical Oncology Department, Ospedale Padre Antero Micone, 16153 Genoa, Italy
5
Department of Medical, Surgical and Experimental Sciences, University of Sassari, Via Roma 151, 07100 Sassari, Italy
6
Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
7
Department of Electronics, Information, and Bioengineering, Polytechnic University of Milan, Piazza Leonardo da Vinci 32, 20133 Milan, Italy
8
Department of Public Health, University of Naples Federico II, 80138 Naples, Italy
9
UO Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(5), 2625; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22052625
Submission received: 14 February 2021 / Revised: 28 February 2021 / Accepted: 1 March 2021 / Published: 5 March 2021
(This article belongs to the Special Issue The New Molecular Strategies under Development in Thoracic Oncology)

Abstract

:
In the scenario of systemic treatment for advanced non-small cell lung cancer (NSCLC) patients, one of the most relevant breakthroughs is represented by targeted therapies. Throughout the last years, inhibitors of the epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), c-Ros oncogene 1 (ROS1), and V-raf murine sarcoma viral oncogene homolog B (BRAF) have been approved and are currently used in clinical practice. However, other promising molecular drivers are rapidly emerging as therapeutic targets. This review aims to cover the molecular alterations with a potential clinical impact in NSCLC, including amplifications or mutations of the mesenchymal–epithelial transition factor (MET), fusions of rearranged during transfection (RET), rearrangements of the neurotrophic tyrosine kinase (NTRK) genes, mutations of the Kirsten rat sarcoma viral oncogene (KRAS) and phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha (PIK3CA), as well as amplifications or mutations of human epidermal growth factor receptor 2 (HER2). Additionally, we summarized the current status of targeted agents under investigation for such alterations. This revision of the current literature on emerging molecular targets is needed as the evolving knowledge on novel actionable oncogenic drivers and targeted agents is expected to increase the proportion of patients who will benefit from tailored therapeutic approaches.

1. Introduction

Since the discovery of actionable molecular alterations, the therapeutic approach of advanced non-small cell lung cancer (NSCLC) patients has been constantly evolving throughout the years. As the current paramount of molecularly driven treatment of NSCLC is represented by mutations of the epidermal growth factor receptor (EGFR) [1], rearrangements of the anaplastic lymphoma kinase (ALK) [2] or c-Ros oncogene 1 (ROS1) [3], as well as mutations of the v-Raf murine sarcoma viral oncogene homolog B (BRAF) [4], the identification of these genetic alterations is considered pivotal in current clinical practice worldwide. Nonetheless, other oncogenic drivers with therapeutic relevance have emerged more recently, and clinically active inhibitors have been approved or are under development.
This review aims to summarize the current state of the art and future directions of novel emerging therapeutic targets for NSCLC, with mention to the involved molecular pathways and the potential therapeutic strategies assessed in clinical studies. We performed a comprehensive literature search to collect relevant data on PubMed for published articles and the most relevant international conferences involving lung cancer from the American Association for Cancer Research (AACR), American Society of Clinical Oncology (ASCO), European Society for Medical Oncology (ESMO), and World Conference on Lung Cancer (WCLC), looking for the most updated and complete information.

2. MET

2.1. Epidemiology

Mesenchymal–epithelial transition factor (MET)-activating alterations, including overexpression (15–70%), amplification (2–5%), and exon 14 (METex14) skipping mutations (3–4%), are oncogenic drivers in 5–9% of newly diagnosed non-squamous NSCLC [5,6]. MET amplification is also responsible for the 10–20% of the acquired resistance, especially to EGFR inhibitors, but also to ALK inhibitors, although METex14 mutations are also emerging as a resistance mechanism [7,8]. Alterations of METex14 are generally mutually exclusive with other primary oncogenic drivers, except for MET amplifications and copy number variants [9].
Several studies showed that MET-positive patients are more frequently Caucasian, elderly (median age >70 years), female, never-smokers, and at an earlier stage compared with other oncogene-addicted NSCLC patients [10,11,12]. Alterations of MET are generally associated with aggressive disease, resistance to anticancer therapies, and a poor prognosis when not treated with MET inhibitors [11,13,14].
These aberrations have been identified mostly in the adenocarcinoma histology, especially with sarcomatoid and adenosquamous features (10–20%) [10,11]. In a retrospective analysis on radiological features of METex14-mutated NSCLC patients, the primary tumor seemed to present as solid and peripheral masses with a high frequency of multifocal and extrathoracic metastases, mainly to the bone, brain, and adrenal glands [15].

2.2. Molecular Pathway

The c-MET gene is a proto-oncogene located at chromosome 7q21-q31, which encodes for a heterodimer receptor tyrosine kinase (RTK), also known as hepatocyte growth factor receptor (HGFR), with extracellular, transmembrane, juxtamembrane, and kinase domains [9].
The binding of MET to its ligand HGF leads to its homodimerization with subsequent auto-phosphorylation and activation of the intracellular tyrosine kinase domains. The activation of c-MET stimulates many downstream signaling pathways, including mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK), phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT), mammalian target of rapamycin (mTOR), and Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathways [9,16]. These pathways are involved in cell survival, proliferation, migration, invasion, angiogenesis, and the epithelial-to-mesenchymal transition.
The MET exon 14 encodes the juxtamembrane domain containing the tyrosine 1003 (Y1003), which is the binding site of a ubiquitin protein (Cbl) that regulates ubiquitin-mediated c-MET receptor degradation. The METex14 skipping mutations alter the splicing process, producing a MET variant that lacks Y1003, which consequently cannot bind Cbl. This alteration results in decreased degradation and subsequent ligand-independent MET activation [17].

2.3. Diagnostic Methodology

Many diagnostic techniques can be used for the detection of MET alterations, both as gene aberrations and protein expression. Immunohistochemistry (IHC) has been used to detect MET overexpression and in past clinical trials to select and stratify patients. However, the level of MET protein overexpression and its activity has been observed to be poorly correlated [18]. For this reason, MET IHC assessment has been replaced with other techniques. Gene amplifications are mostly detected by fluorescence in situ hybridization (FISH), but also DNA/RNA sequencing techniques are employed. However, there is no consensus on the definition of MET amplification since different studies have proposed various scores and different cut-offs [16]. METex14 mutation can be detected by DNA sequencing or RNA-based sequencing techniques, including next-generation sequencing (NGS), which is the primary detection method, NanoString, and quantitative real-time polymerase chain reaction (qRT-PCR) [16,18].

2.4. Therapeutic Implications

The two most clinically relevant MET alterations are METex14 skipping mutations as the primary oncogenic driver and MET amplification as acquired resistance to tyrosine kinase inhibitors (TKI). In these settings, many anti-MET drugs with different mechanisms of action, either as single agents or combinations, have been developed and are currently under investigation. They are classified into MET TKIs and monoclonal antibodies (anti-MET antibodies and anti-MET antibody-drug conjugates) [11].
Since data on MET-inhibitors are rapidly expanding, here we mainly discuss the major therapeutic strategies, referring for details to dedicated reviews and clinical trials’ results [5,9,10,11].

2.4.1. MET as Primary Oncogenic Driver

MET TKIs are the principal MET-targeted drugs for METex14 mutations and they are divided into non-selective (multi-targeted) TKIs and the more efficient selective TKIs. Among the first ones, the most studied TKI with positive results was crizotinib, whereas the second ones (e.g., capmatinib, tepotinib, savolitinib) have recently provided encouraging data [6] (Table 1). Due to the great number of MET inhibitors currently under investigation, in this review we focused on the Phase II–III trials of selective TKIs.

Crizotinib

In 2018, crizotinib received breakthrough therapy designation from the US Food and Drug Administration (FDA) based on its efficacy observed in the expansion cohort of the PROFILE 1001 trial on METex14-mutated advanced NSCLC patients [19,20]. More recently, the updated data reported an overall response rate (ORR) of 32% and a median progression-free survival (mPFS) of 7.3 months [21]. Many Phase I–II (e.g., AcSè, METROS) and retrospective analyses confirmed the reported activity and survival results [16,22,23,24]. In these studies, crizotinib reported similar results in MET-amplified patients. Therefore, crizotinib has been recommended for both METex14-mutated and MET-amplified NSCLC patients according to international guidelines [25].

Capmatinib

Capmatinib is the first FDA-approved selective MET-inhibitor in METex14-mutated NSCLC patients and its use is currently suggested in the first-line setting [25,26]. The approval (May 2020) was based on the efficacy reported by the multi-cohorts Phase II GEOMETRY mono-1 trial [27]. In this study, capmatinib was assessed as first- and subsequent-line in MET-dysregulated advanced NSCLC patients, showing a promising antitumor activity among METex14-mutated patients, especially untreated ones and those with brain metastases. In MET-amplified patients, the efficacy was higher in tumors with a high gene copy number. The randomized Phase III GeoMETry-III trial (NCT04427072) is currently ongoing to evaluate capmatinib compared to docetaxel in pre-treated NSCLC patients. The primary endpoint is mPFS and secondary endpoints included median overall survival (mOS), ORR, and median duration of response (mDOR). Ongoing trials are assessing capmatinib in METex14-mutated patients progressing to MET-inhibitors (NCT02750215) or systemic therapy for advanced disease (NCT04427072).

Tepotinib

The ongoing Phase II VISION study is evaluating the activity of tepotinib in NSCLC patients harboring METex14 mutations detected by liquid and tissue biopsies. Preliminary data were presented at the 2019 ASCO annual meeting and the final results have been recently published [28,29]: tepotinib has shown promising activity and long responses across treatment lines, including patients with brain metastases.
Based on the preliminary data, in September 2019 tepotinib obtained the FDA breakthrough therapy designation for metastatic METex14-mutated NSCLC patients that progressed after platinum-based chemotherapy [30].

Savolitinib

Savolitinib, a novel potent and selective MET-inhibitor that is under evaluation in a Phase II study on METex14-mutated NSCLC (NCT02897479), has demonstrated promising anti-tumor activity and a durable response in a preliminary analysis [31].

Other MET-Inhibitors

Other MET-TKIs are currently under evaluation in Phase II trials, including cabozantinib (NCT03911193, NCT01639508), merestinib (NCT02920996), bozitinib (NCT03175224, NCT04258033), and Glumetinib (NCT04270591) [9,10].

2.4.2. MET as Secondary Acquired Resistance

Amplification of MET is the main resistance mechanism to EGFR-TKIs in NSCLC patients, both for the first- and second-generations (50–60%) and for the third-generation (15–20%) [17]. Several ongoing trials are investigating the role of MET-TKIs as a single agent or in combination with EGFR-TKIs, to overcome the acquired resistance to anti-EGFR therapies (Table 2). Therapeutic strategies included the addition of a MET-inhibitor to the previous EGFR-TKI. In the case of first- and second-generation EGFR-TKI, the exon 20 p.T790M mutation had not been present at disease progression. Capmatinib, cabozantinib, and tepotinib in combination with EGFR-TKIs have already yielded promising antitumor activity in EGFR-mutated, MET-amplified NSCLC patients [16,18,32].

2.4.3. MET Alterations and Immunotherapy

The MET activation has shown to induce programmed cell-death protein-1–ligand-1 (PD-L1) expression, contributing to immune suppression and evasion [33]. Several retrospective analyses have reported controversial results on the predictive value of MET alterations to immunotherapy [10,17]. In the IMMUNOTARGET study, 36 MET-positive NSCLC patients receiving immunotherapy experienced limited activity (ORR 16%) and survival outcomes (mPFS and mOS of 3.4 and 18.4 months, respectively), similar to other analyses [34,35]. However, durable responses were also observed by other studies in the same setting [36,37].
Immunotherapy generally seemed to have less anti-tumor activity in MET-positive NSCLC patients compared with TKIs and chemotherapy. Moreover, PD-L1 and tumor mutational burden (TMB) seemed not to predict response to immunotherapy in these patients [9]. Therefore, immunotherapy should be considered only after failure of other effective treatments, although the role of immune-based combinations in this setting needs further investigation [17]. In this context, several Phase I–II trials on the combination of MET-TKIs and immunotherapy are ongoing (e.g., NCT02323126, NCT03468985).

3. RET

3.1. Epidemiology

Alterations of the rearranged during transfection (RET) gene in NSCLC are detected in approximately 2% of patients [38,39,40]. Overall, RET aberrations are more represented in adenocarcinoma histology. Patients with lung cancer and RET involvement are frequently never-smokers and young. These neoplasms can present a solid pattern with signet rings or might otherwise be characterized by a lepidic pattern. The primary tumor is often small but with early lymph node involvement. Alterations of RET and other NSCLC driver aberrations were initially thought to be mutually exclusive, but there are consistent data on the presence of concomitant gene alterations in a small percentage of cases [40,41].

3.2. Molecular Pathway

The RET gene is a proto-oncogene located on chromosome 10q11.2 that encodes for a transmembrane receptor tyrosine kinase (RTK), consisting of an extracellular domain, a hydrophobic transmembrane domain, and an intracellular domain, including a juxtamembrane portion, a tyrosine kinase domain, and carboxy-terminal tail. Activation of RET is given from the interaction of the glial cell line-derived neurotrophic factor (GDNF) family receptor α (GFRα) co-receptor with ligands belonging to the family of neurotrophic factors, including GDNF [42], neurturin (NRTN) [43], persephin (PSPN) [44], and artemin (ARTN) [45]. The physiological ligands of RET are part of the growth factors of the glia; indeed, RET is physiologically present in the neuroectodermal tissues and is crucial for the development of the fetus, in particular for the genesis of the enteric parasympathetic nervous system [46]. The interaction between the receptor and its ligands ultimately leads to the formation of homodimers and the phosphorylation of tyrosine residues of the kinase domain. The phosphorylation of the SH domain activates downstream signaling pathways, such as RAS/MAPK/ERK, PI3K/AKT, and JAK/STATs, which, in turn, lead to signal transduction for cell proliferation, differentiation, and migration.
To date, several fusion partner genes of RET have been discovered, the most frequent being the rearrangement with kinesin family member 5B gene (KIF5B-RET) of which there are seven variants, coiled-coil domain containing 6 (CCDC6–RET), nuclear receptor coactivator 4 (NCOA4-RET), and tripartite motif-containing 33 (TRIM33–RET) [47]. Irrespective of the gene partner and the breakpoint, RET fusions lead to the formation of a new protein that includes the coiled-coil domain of the partner gene and RET intracellular kinase domain, which always retains the tyrosine kinase activity. The coil-coiled domain of the partner gene leads to a ligand-independent RET dimerization, resulting in its constitutive activation.

3.3. Diagnostic Methodology

The diagnostic methods to detect RET fusions are still under debate. More specifically, IHC has demonstrated a low specificity in detecting RET rearrangements [39,48,49] and it is not considered applicable in clinical practice. Conversely, high sensitivity and specificity are guaranteed by NGS, FISH, and qRT-PCR techniques. The NGS approach, both whole genome and whole transcriptome sequencing, represents the best technology for discovering novel RET rearrangements, although it is still significantly expensive; moreover, it necessitates computational expertise and infrastructures, preventing its application in a diagnostic context. Currently, the standard for diagnosing RET fusions is represented by FISH [49]. This technique is highly sensitive and able to detect fusions independently of the partner genes, although it cannot define which fusion gene is involved. Nonetheless, it requires experienced personnel for data interpretation, and, to date, there is presently no agreement on the cutoff to be used to define the positivity of RET. Conversely, RT-PCR has the great advantage over FISH of identifying the fusion partner gene. Besides, it can provide a diagnosis even on cytological samples, compared to FISH that needs histological samples [50]. Unlike FISH, a possible limitation is that RT-PCR cannot trace unknown or novel fusion partners, resulting in an underestimation of the prevalence of RET rearrangements [51]. Consequently, these two techniques have often been combined as screening methods.

3.4. Therapeutic Implications

Previous studies have shown that RET fusions are associated with a lower expression of thymidylate synthase [41], which is linked to high sensitivity to treatment with pemetrexed, with an ORR reaching up to 40%. In the last year, two targeted therapies have shown an important activity in these patients, surpassing the results achieved by other TKIs, so far. Indeed, selpercatinib and pralsetinib (Blue 667) have received FDA approval for the treatment of patients with RET fusion in NSCLC, due to their activity and efficacy. Previously, vandetanib, lenvatinib, sorafenib, cabozantinib, and alectinib, reached in small studies an ORR ranging from 16 to 50% and an mPFS that did not exceed 6 months [52,53,54,55]. LIBRETTO-001, a Phase III trial of selpercatinib for the treatment of NSCLC patients, showed an ORR of 64% in pretreated patients, with an mPFS of 17.5 months. The trial enrolled also previously untreated patients. In the subgroup of 39 patients who received selpercatinib as first-line treatment, the ORR was 85% [56]. Pralsetinib is another drug approved in 2020 by the FDA for the treatment of NSCLC patients based on the results of the phase I/II ARROW trial. Pralsetinib demonstrated an ORR of 57% in pretreated patients while it reached 70% in previously untreated patients, a response that lasted at least 6 months in 80% of pretreated patients and 58% of naïve patients [57] (Table 3).

4. NTRK

4.1. Epidemiology

Alterations of the neurotrophic tyrosine kinase (NTRK) genes (i.e., NTRK1, NTRK2, and NTRK3) are rare in NSCLC, representing less than 1% (about 0.1–0.6%) of the NSCLC population [5,58,59]. Although the proportion is very small, the global incidence of NSCLC patients is high, making these relatively few cases a relevant number of patients in absolute terms. Given the rarity and the relatively recent discovery of this target, any solid data on morphological and clinical characteristics of NSCLC with NTRK involvement are still lacking.
The largest study on NSCLC retrospectively included 4872 cases and identified only 11 cases with NTRK1-3 fusions. Of these cases, nine were adenocarcinoma, including two mucinous and one with neuroendocrine features, while one was a squamous cell carcinoma (SCC), and one a large-cell neuroendocrine carcinoma [59].
High expression of the protein encoded by NTRK2 (TrkB) and of its ligand, namely brain-derived neurotrophic factor (BDNF), are the only proteins found to be correlated with a higher prevalence of vascular invasion, lymph nodes metastases, and advanced stage in NSCLC patients. These factors resulted in a poorer prognosis for this population [60,61].
The other relevant clinical characteristic that has emerged is that the majority of NTRK-driven NSCLC patients were never-smoker (about 80%), as it is usually observed for other kinase fusion-positive NSCLCs [62].
Interestingly, different studies highlighted that NTRK fusions seem to be almost mutually exclusive with alterations in other known genes, such as ALK, ROS-1, MET, and RET [63,64].

4.2. Molecular Pathway

The NTRK genes encode the TrkA, TrkB, and TrkC transmembrane glycoproteins, respectively, that together with their natural ligands, which are the nerve growth factor (NGF), BDNF, neurotrophin-3 (NT-3), and NT-4, are involved in the physiological development and function of the central and peripheral nervous systems [59,60,61]. Fusions of NTRK genes lead to overexpression of Trk proteins and, therefore, to the constitutive activation of downstream signaling pathways such as RAS/MAPK, PI3K/AKT, and PLC-γ, responsible for cancer cells transformation, proliferation, and survival [5,61]. The most commonly detected fusions are ETS Variant Transcription Factor 6 (ETV6)-NTRK3 and Echinoderm Microtubule Associated Protein Like 4 (EML4)-NTRK3, although more than 50 other fusion partners are currently known [65].
Interestingly, another role for one of these transmembrane proteins has been recently found. Indeed, preclinical studies have shown that the increased expression of TrkB promotes the suppression of E-cadherin expression and enhances the activity of the matrix metalloproteinase-2 (MMP-2) in lung SCC cells, promoting cancer aggressiveness [60,61].

4.3. Diagnostic Methodology

The NTRK fusions can be studied with different methods, among which IHC is one of the most widespread used and characterized by high sensitivity (95 to 100%) and specificity (93% to 100%); although these data derive from very small studies, this technique can be considered a good screening tool as of today, given the rarity of NTRK gene alterations [58,62].
Another relevant diagnostic tool is represented by FISH, which may be helpful when the histologic tumor type is known to frequently harbor an NTRK fusion, although this may not be the best option in NSCLC due to its aforementioned low prevalence [59,62]. Nowadays, the employment of newly approved targeted therapies may be either approved for patients with an NTRK gene fusion diagnosed by NGS techniques or within clinical trials. IHC testing can be accepted but needs to be validated with another diagnostic method, thus resulting in a more time-consuming procedure for each patient. Thus, the NGS approach may be proposed either as front-line or after positivity at the IHC screening as it is recommended in the latest ESMO guidelines [66].

4.4. Therapeutic Implications

The discovery of actionable NTRK gene fusions represented a revolution in oncology. The first-generation TRK inhibitors entrectinib and larotrectinib received, in fact, an agnostic approval by the FDA for patients with NTRK fusion-positive solid tumors because of their impressive results in Phase I/II trials [5,59].
Entrectinib is a TRKA, TRKB, TRKC, ROS1, and ALK multi-inhibitor, and data from three Phase I/II trials (ALKA-372-001, STARTRK-1, and STARTRK-2) reported an ORR for NSCLC patients of 70–75% with 10% of complete responses (CR), an mDOR of 12.9 months, and an mOS of 23.9 months (Table 4) [67,68,69]. Larotrectinib, by contrast, is a pan-TRK inhibitor and a recently published pooled analysis of three Phase I/II trials showed, among NSCLC patients, an ORR of 75%, an mPFS of 28.3 months, and an mOS of 44.4 months [70].
These two drugs are both very effective and very well tolerated with few side effects. The main difference is the central nervous system (CNS) penetration as entrectinib can cross more effectively the blood–brain barrier. As a matter of fact, entrectinib has proved its efficacy in patients with brain metastases, although it has also shown some CNS side-effects, such as dizziness [67,68,69].
Across time, as in many other targeted therapies, the tumor can develop acquired resistance, but new generations of TRK inhibitors, such as taletrectinib (TPX-0005), selirectinib (LOXO-195), and repotrectinib, are currently under evaluation in ongoing Phase I/II clinical trials [5,59].

5. KRAS

5.1. Epidemiology

Kirsten Rat Sarcoma viral oncogene (KRAS) mutations are among the most frequently detected oncogenic drivers in NSCLC. These mutations were identified in lung cancer almost three decades ago and have been traditionally associated with a poor prognosis compared to KRAS wild type tumors [71]. KRAS mutations were found in 26% of NSCLC, almost exclusively in adenocarcinomas and in the smoker population [72]. Only a minority of never smokers (6%) harbors the KRAS-mutant NSCLC. Smoking history significantly increases the chance to detect KRAS mutation in lung cancer, regardless of pack-years of smoking [73].

5.2. Molecular Pathway

RAS genes, including KRAS, are proto-oncogenes encoding intracellular guanine nucleotide-binding proteins that belong to the GTPase family. RAS proteins are composed of a catalytic domain called “G domain”, which binds guanine nucleotides and activates signaling, and a hypervariable region (HVR), which determinates where the RAS proteins are localized on the cell membrane to perform their signaling function. The downstream signaling depends on the RAS bound state: the guanosine triphosphate (GTP)-bound state is the active form while the guanosine diphosphate (GDP)-bound state is the inactive form [74]. GTP/GDP cycling is regulated by guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs); GEFs promote RAS activation by binding RAS and causing GDP separation. Otherwise, GAPs stimulate the intrinsic GTP hydrolysis activity of RAS to increase inactive RAS–GDP forming [75]. The RAS–GTP complex activates multiple signaling cascades in response to extracellular signals. Some of these pathways include RAF/MEK/ERK, PI3K/AKT/mTOR, RalA/B, and TIAM1/RAC1, which regulate cell proliferation, differentiation, and apoptosis. Most RAS mutations usually involve exons 2 and 3, impairing the conversion from GTP-bound to GDP-bound state and constitutively activating the downstream signaling. The most frequent mutations are the nucleotide substitutions in 12 codons of KRAS exon 2 p.G12C, p. G12D, and p.G12V [74].

5.3. Diagnostic Methodology

Currently, the evaluation of KRAS gene alterations is strongly recommended in patients with colon–rectal cancer or NSCLC eligible for anti-EGFR therapy [25]. Many studies have indeed demonstrated that KRAS mutations are negative biomarkers of response to anti-EGFR. This mechanism of resistance derives from the persistent stimulation of the EGFR/RAS/RAF/ERK/MEK pathway when KRAS is mutated, regardless of the mutational status of EGFR. The KRAS testing is usually performed on tumor tissue, rather than from secondary lesions in case of metastatic disease. There are more than 60 methods currently available for KRAS testing, including sequencing, high-resolution melting analysis (HRM), single-strand conformation polymorphism (SSCP), denaturing gradient gel electrophoresis (DGGE), denaturing high-performance liquid chromatography (DHPLC), array/strip analysis, and allele-specific PCR [76].

5.4. Therapeutic Implications

Over the past few years, several efforts have been done to find a predictive role of KRAS alterations. While data from chemotherapy response are conflicting, some trials have shown a correlation between KRAS mutations and the benefit from immune checkpoint inhibitors [77]. A meta-analysis by Kim et al. demonstrated that immune checkpoint inhibitors therapy significantly improved OS in patients with KRAS-mutant NSCLC but not in those with the KRAS wild-type tumor [78]. The biological rationale is probably related to an increased expression of PD-L1 and high tumor immunogenicity [79]. However, co-occurring serine/threonine kinase (STK11) and KRAS mutations seem to be associated with PD-1 inhibitor resistance [80].
To date, the standard of care for KRAS-driven tumors is the same as that for non-oncogene addicted NSCLC since no targeted therapies are approved. Different strategies to directly target KRAS or its downstream effectors have been developed, either as a single agent or in combination with other TKIs, chemotherapy, and immunotherapy (Table 5).
Several preclinical and clinical trials have investigated the inhibition of multiple KRAS pathways but most of them failed in giving significant survival or response improvements [74].
Recent developments of novel targeted therapies against KRAS exon 2 p.G12C have shown encouraging results. These molecules irreversibly bind to KRAS exon 2 p.G12C in the GDP-bound state, preventing the conversion to the active form and impairing the downstream signaling [79]. Preliminary data have demonstrated anti-tumor activity with durable responses and a favorable safety profile [81,82]. Currently, a multicenter Phase III clinical trial of sotorasib (CODEBREAK200) in previously treated advanced KRAS-mutant NSCLC is ongoing (NCT04303780).
An additional therapeutic strategy has been explored with the use of cyclin-dependent kinase (CDK) 4/6 inhibitors, as a synthetic lethal interaction between KRAS oncogenes and CDK4 in NSCLC [83]. An ongoing Phase III clinical trial (JUNIPER) is evaluating the efficacy and safety of abemaciclib compared to erlotinib in previously treated patients with advanced KRAS-mutated NSCLC. The study did not meet its primary endpoint of OS but demonstrated an improvement in PFS and ORR [84].

6. PIK3CA

6.1. Epidemiology

The phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha (PIK3CA) gene encodes for the catalytic subunit alpha of the phosphoinositide 3-kinase (PI3K) protein. Mutations of PIK3CA in lung cancer are uncommon, being prevalently detected in SCC histology, while its amplification is conversely frequently found [85]. The most common mutations are reported in exon 9 and 20. In tumor samples from 1144 NSCLC patients, PIK3CA mutations had an incidence of 3.7%, raised to 8.9% if counting only SCC [86], while in another cohort of 1117 NSCLCs, they were detected in 3.9% of SCCs and 2.7% of adenocarcinomas [87]. Nevertheless, PI3K/AKT/mTOR signaling is one of the main activated intracellular downstream pathways in different cancer types. The prognostic value of PIK3CA mutations is still partially undefined since survival analyses from different studies were discordant [86,87]. Nevertheless, a recent meta-analysis correlated the presence of PIK3CA mutations with worse OS and PFS in patients with NSCLC [88]. This uncertainty is probably related to the co-occurrence of alterations other than those involving PI3K; a genomic sequencing of specimens from early-stage NSCLC revealed PIK3CA as the most frequently mutated gene in co-existence with EGFR and KRAS mutations [89]. Patients harboring PIK3CA mutations are more commonly current or former smokers, although they were also observed in never smokers [87]. Notably, PIK3CA mutations are more frequently detected in metastases than in the primary tumor [90]. Thus, PIK3CA-mutated NSCLC represents a clinically and genetically heterogeneous subgroup [91].

6.2. Molecular Pathway

The PI3Ks are a family of lipid kinases able to phosphorylate the 3’-OH group of phosphatidylinositols and phosphoinositides on the cell membrane. These kinases are heterodimers with a catalytic (p110) and regulatory (p85) subunit, grouped in classes; class IA proteins, mainly related to human cancer, are usually activated by growth factor receptors, such as EGFR, insulin growth factor 1-receptor (IGF1-R), and HER2/neu. Such receptors represent the upstream signal of PI3K, leading to the activation of the catalytic domain of p110, which phosphorylates the phosphatidylinositol bisphosphate (PIP2) to phosphatidylinositol triphosphate (PIP3); subsequently, PIP3 locates AKT to the plasma membrane by binding to the phosphorylated lipid products [92]. Phosphatase and tensin homologue deleted on chromosome 10 (PTEN) acts as an inverse regulator through PIP2 and PIP2 dephosphorylation. AKT is a serine/threonine protein able to phosphorylate and consequently activate/inactivate numerous downstream pathways involving cytoplasmic and nuclear substrates that regulate apoptosis, cell cycle, cell survival, and proliferation, among which nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) transcription factor is one of the most important [92]. mTOR is a serine/threonine kinase complexed with other proteins to form the mTORCH complexes. mTORC2 promotes the activation of AKT via phosphorylation at serine 473 [91], while mTORC1 phosphorylates the p70S6 kinase (S6K1) and the eukaryotic initiation factor 4E binding protein 1 (4EBP1), leading to increased protein synthesis and cell growth [93].

6.3. Diagnostic Methodology

Currently, the evaluation of PIK3CA gene alterations is not recommended in clinical practice since no targeting drugs have been approved to date for lung cancer treatment. Activating mutations in PIK3CA can be detected in tissue or plasma specimens. Diagnostic testing within clinical trials is usually performed using NGS (see studies in Table 6).

6.4. Therapeutic Implications

PI3K inhibitors can be classified into pan-PI3K inhibitors [94,95,96,97,98,99,100,101,102], PI3K and mTOR inhibitors [103], and selective PI3K inhibitors [104]. The molecular downstream pathways can also be blocked by single inhibition of AKT and mTOR.
Table 6 reports the main characteristics of clinical studies investigating the role of PI3K inhibitors in the treatment of advanced stage lung cancer. Several other PI3K-targeting agents, not reported in this review, have been tested at pre-clinical or clinical level, with inconsistent results.

7. HER2

7.1. Epidemiology

Three main alterations involving the human epidermal growth factor receptor 2 (HER2) can be identified: HER2 gene mutations, HER2 gene amplification, and HER2 protein overexpression.

7.1.1. HER2 Mutations

Mutations of HER2, mostly represented by exon 20 insertion, account for a minor number of cases of adenocarcinomas of the lung (2 to 4%) [105] and are generally associated with a particular clinical phenotype presentation. Indeed, similarly to EGFR exon 20 insertions, they are usually found in younger patients, never smokers, and in patients with a smaller tumor size. Conversely, no relationship with sex, lymph nodes involvement, or tumor stage has been reported, so far. Additionally, unlike EGFR mutations, the prevalence of HER2 insertions appears to be comparable between Caucasian and Asian populations. Mutations in HER2 appear to be mutually exclusive with other drivers of genetic alterations. Furthermore, HER2 represents an independent poor prognostic factor and, similarly to EGFR insertions, is intrinsically resistant to the currently available therapies [106,107].

7.1.2. HER2 Amplification and Protein Expression

Up to now, the biologic relationship of HER2 mutations, amplifications, and protein overexpression has not definitively been elucidated. HER2 amplification and HER2 overexpression (defined as a high level (3+) by IHC) have been detected in 2% to 5% and 2% to 4% of lung cancer, respectively. This confusion forcefully discloses during patient selection phase of clinical trials testing HER2-targeted therapies for NSCLC [108].
On this matter, in 2015 Li et al. assessed the presence of HER2 alterations on tumor samples from 175 patients with recurrent/stage IV lung adenocarcinomas. HER2 amplification and HER2 mutations were detected in 5/175 and 4/148 patients (27 cases had no evaluable specimen), respectively, including three patients with an identical 12-base-pair insertion (p.A775_G776insYVMA; c.2324_2325ins12) in exon 20 and one with an unspecified 9-base-pair insertion in exon 20. Forty-six patients showed a polysomy (HER2 copy number ≥ 4 but HER2-to-CEP17 ratio <2). No match was found between four mutations and amplifications and no HER2 overexpression was identified [109]. Other studies confirmed no overlapping between HER2 mutations and HER2 amplification in NSCLC, enhancing the assumption that they represent two different cancerogenic entities [105,109].

7.2. Molecular Pathway

HER2 belongs to an RTK family that includes EGFR (ERBB1), HER2 (ERBB2/NEU), HER3 (ERBB3), and HER4 (ERBB4). HER2 is a 185 kDa transmembrane glycoprotein encoded by the ERBB2 gene located at chromosome 17q. As mentioned above, dysregulation of HER2 in NSCLC can be caused by mutations, amplification, and protein overexpression [110,111]. The amplification of HER2 promotes tumorigenesis and is involved in the pathogenesis of several human cancers. In contrast to EGFR, no ligand has yet been recognized for HER2. Still, HER2 seems to be the main involved partner in the dimerization of all ERBB family components [112]. After the ligand binds to the extracellular domain of the receptor, HER2 undergoes heterodimerization, inducing a HER2 tyrosine kinase activation through phosphorylation of the intracellular tyrosine kinase domain, leading to activate several downstream signaling pathways. These include the RAS–RAF–MEK–ERK (MAPK) and the PI3K–AKT–mTOR pathways, which are involved in cell survival, proliferation, and apoptosis [113].

7.3. Diagnostic Methodology

Currently, there are different methods to diagnose HER2 overexpression, including IHC, Western blot, and enzyme-linked immunosorbent assay (ELISA), whereas, FISH, silver in situ hybridization (SISH), chromogenic in situ hybridization (CISH), PCR, Southern blot, and NGS [114] are employed to detect HER2 amplification [115]. Finally, HER2 mutations are detected by NGS, Sanger sequencing, qRT-PCR, and matrix-assisted laser desorption ionization–time of flight mass spectrometry (MALDI-TOF MS) [116]. Approximately 80% of HER2 evaluations start with IHC as a screening test. Among these tests, IHC and FISH constitute the current standard to assess HER2 expression and HER2 amplification, respectively. While IHC is a quick and cost-effective test [117], FISH is a highly sensitive and specific assay [118]. Finally, sequencing methods are probably the most used approaches for detecting HER2 mutation [119]. However, except for qRT-PCR, all the other approaches can also detect the exact amino acid positions.

7.4. Therapeutic Implications

Among new generation anti-HER2 for HER2 mutation the most promising therapeutic strategies are reported below and in Table 7.
In the ASCO 2020 Meeting, results from the DESTINY-Lung01 multicenter Phase II trial were presented. Forty-two patients diagnosed with relapsed/refractory NSCLC HER2 mutation were treated with an anti-HER2 antibody conjugate called fam-trastuzumab deruxtecan-nxki (T-DXd; Enhertu). Promising results reported an ORR of 61.9% (1 CR, 25 PR and 12 SD) and an mPFS of 14.0 months (95% CI, 6.4–14.0), with a not reached mDOR (95% CI, 5.3–not evaluable) and OS. The most common serious (Grade ≥ 3) drug-related adverse events (AEs) included neutropenia (26.2%) and anemia (16.7%), with particular attention to an AE of special interest such as interstitial lung disease in 11.9% with G2 severity [120]. Based on these results, a breakthrough therapy designation has been granted from the FDA for the T-DXd drug in HER2-mutant NSCLC and gastric cancer [121].
Poziotinib is an oral, irreversible pan-HER TKI, initially investigated in the Asian population. In the ESMO 2020 Congress, ZENTITH20-2, a multicenter phase II trial, reported results from a cohort of 90 HER2 mutated patients. ORR was 35.1% (95% CI: 24.4–47.1%) and 27.8% (95% CI: 18.9–38.2%) in 74 evaluable patients and in all 90 patients, respectively. Median PFS and mDORe were 5.5 months (range: 0.03–13.1+ months) and 5.1 months (range: 1–12.3+ months), respectively. Patients with brain metastases obtained an ORR of 28.6% [122]. The most common treatment-related grade ≥3 AEs were rash (30%), diarrhea (26%), and mucosal inflammation (14%). Results from the first expanded access program of 30 EGFR/HER2 insertion reported a higher ORR for HER2 insertion compared to EGFR insertion patients (50% vs. 19%), with no differences in mPFS (5.6 months, CI95% 3.6–7.1 months) [123].
Pyrotinib is another oral, irreversible pan-HER TKI against HER1, HER2, and HER4 for HER2+ (immunochemistry expression) tested in NSCLC [124]. In 2019, Gao et al. published the first results of an open-label, multicenter, single-arm Phase II study. Sixty pretreated NSCLC patients with HER2 insertion were enrolled, whereas patients with active brain metastases or those already treated with an anti-HER2 targeted therapy were excluded. The ORR was 31.7% (95% CI; 20.3–45.0), while the PFS was 6.8 months (95% CI 4.1–8.3). Diarrhea was reported as the only treatment-related grade 3 AE, occurring in ≥2 patients [125].
Among the new-generation TKIs, mobocertinib (TAK788) is a small-molecule oral TKI designed to selectively target both EGFR insertion and HER2 mutation. Exciting results from an ongoing Phase I/II NCT02716116 trial including both EGFR insertion and HER2 mutation in different cohorts pushed the FDA for a breakthrough therapy designation for EGFR insertion. Results from ongoing studies on HER-mutated patient cohorts are awaited [126].
Lastly, in 2019, results from the multicenter single-arm Phase II NICHE trial were published by Dziadziuszko et al., testing afatinib, an EGFR inhibitor, in 13 NSCLC patients pre-treated with platinum-based CHT harboring HER2 mutations. Afatinib did not show the expected disease control, with a median PFS of 15.9 weeks (95% CI: 6.0–35.4) and OS of 56 weeks (95% CI; 16.3 weeks –NR). Moreover, severe drug-related AEs were reported: dyspnea (15.3%), acute renal injury (7.6%), and mucositis (7.6%) [127].
Several other drugs, such as trastuzumab and emtansine, have also demonstrated a role in HER2-altered NSCLC, but none of them has demonstrated a practical clinical application so far.
Therefore, the definition of “HER2+ NSCLC” is insufficient, as it does not exhaustively distinguish the complexity of this gene alterations. Indeed, the failure of many clinical trials aimed at targeting HER2 may have been derived from considering these alterations as a single entity [127,128,129]. Conversely, the DESTINY-Lung01 trial, which distinguished between HER2 overexpression and HER2 mutation in the two different cohorts, is a good example of a successful trial. In Table 7 we reported almost all historical trials that investigated different agents against HER2 alterations.

8. Discussion

Throughout the last decade, the treatment scenario of advanced NSCLC patients has been undergoing a revolution in terms of therapeutic targets. Notably, EGFR represents a paradigm of targetable oncogenic drivers, with a constant increase in terms of translational knowledge and clinical applications; moreover, the subsequent concept of acquired resistance mechanisms, which in turn might become therapeutic targets, is leading the management of EGFR-mutated NSCLC beyond its initial limits [1]. Even though the history of other actionable oncogenic drivers has been shorter and slower compared to EGFR, a similar progress is being achieved, such as the identification of acquired resistance mutations during first- or second-generation ALK inhibitors, and the publication of data suggesting variable sensitivity to the available inhibitors based on the specific reported mutation [130].
In this context, the identification of additional oncogenic drivers that can be exploited in the management of advanced NSCLC patients is expected to significantly increase the proportion of patients receiving targeted therapy (Figure 1).
This is particularly relevant, as the current percentage of patients with clinically actionable drivers in the Caucasian population accounts for 15–20% of non-squamous NSCLC (considering EGFR, ALK, ROS1, and BRAF), and the potential novel emerging targets might further increase this proportion. While the approval of any KRAS exon 2 p.G12C-specific inhibitor would potentially result in a further 10–15% of the patients with non-squamous NSCLC becoming eligible for targeted therapy (the same proportion currently eligible for EGFR inhibitors), the individual impact of each other driver seems more limited. Nonetheless, even if the relative number of patients harboring a specific molecular alteration may represent a little percentage of the whole NSCLC population, testing multiple actionable oncogenic drivers is expected to have a huge impact, because of the relevant absolute numbers of NSCLC patients. Moreover, since patients harboring oncogenic drivers might have an underwhelming clinical benefit from immune checkpoint inhibitors, their identification is decisive to choose the best therapeutic strategy for each patient [34].
An additional subject of discussion is represented by the clinical outcomes observed with targeted therapies. Notably, for several agents, as aforementioned, only Phase I–II data have been generated so far; however, at least for some of these drivers (e.g., MET, RET, NTRK), the observed outcomes have been consistently encouraging, with deep and prolonged objective responses, including intracranial responses. Moreover, the magnitude of clinical benefit appears to be more pronounced if eligible patients receive targeted therapies early, possibly as a first-line treatment.
As a matter of fact, the increased knowledge on actionable genomic targets and the development of effective drugs translates into the necessity to guarantee the widest possible accessibility, both in terms of molecular testing and access to novel compounds.
With regards to molecular testing, the most appropriate assays should be employed, taking into account both the increasing number of evaluated targets and the limited amount of tumor sample available through small biopsies or cell blocks, as well as the time needed to process such samples. The first steps regarding molecular analyses for clinical purposes have been represented for years by single-gene testing: to date, due to the increasing number of tests, such an approach is becoming impractical for NSCLC management. In this setting, the progressive shift towards diagnostic platforms able to analyze multiple genes at once, such as NGS technology, appears to be the next mandatory step for molecular biology laboratories involved in oncology, provided that the necessary resources and skills are optimized, and that the most appropriate sequencing tests are offered to each patient.
Once the tests have been performed and any actionable mutation identified, one additional issue might be represented by accessibility to appropriate targeted agents. Indeed, the rate of approval of novel agents might be different among different countries, especially when different regulatory agencies are involved, such as the FDA and the European Medicines Agency (EMA); as a result, living in different countries may lead to unequal accessibility and disparities. Possible ways to address this issue are represented by clinical trials, which are usually multi-institutional, international studies, as well as expanded access programs, which are designed to provide early access to innovative drugs; based on such information, both approaches should be highly encouraged in cancer centers.
Such is the relevance of properly organized molecular biology tests in oncology and subsequent access to targeted therapies, that several comprehensive cancer centers are focusing on the development of multi-disciplinary Molecular Tumor Boards, designed to provide governance while addressing any disparity in terms of access to oncogenic-driven assays and drugs [131].

9. Conclusions

While acknowledged oncogenic drivers, such as EGFR, ALK, ROS1, and BRAF, have a solid role in the therapeutic algorithms for advanced NSCLC, other actionable targets are progressively being identified, and novel targeted antineoplastic agents are being developed. The most promising novel targets are represented by MET, RET, and NTRK, although other drivers, namely HER2 and PIK3CA, might experience relevant updates in the near future. With regards to KRAS, while this driver has been widely known for years, only recent clinical trials have generated encouraging data, and might eventually result in targeted therapy for a relatively high proportion of NSCLC patients.
Finally, while additional oncogenic targets are being identified, an effort to guarantee accessibility to appropriate molecular testing and novel drugs is a priority in the current setting of NSCLC management.

Author Contributions

Conceptualization, S.E.R., C.G., M.T. and G.R.; investigation, all authors; data curation, all authors; Writing—Original draft preparation, all authors; writing—review and editing, S.E.R., C.G. and M.G.D.B.; visualization, all authors; supervision, S.E.R. and C.G.; funding acquisition, C.G. All authors have read and agreed to the published version of the manuscript.

Funding

No funding was specifically provided for the development of this manuscript. The authors wish to thank the Italian Ministry of Health (5 × 1000 funds Co-2016-02361470; Ricerca Corrente 2018–2020) and Compagnia di San Paolo (2017.0529), that financially support our current research focused on the identification of prognostic and predictive markers for novel antineoplastic treatments.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

C.G. reports honoraria from AstraZeneca, Bristol-Myers-Squibb, Merck-Sharp-Dohme, Boehringer-Ingelheim and Roche, outside the submitted manuscript. M.T. declares travel grants from Roche, Bristol-Myers Squibb, AstraZeneca, Takeda and honoraria as medical writer from Novartis and Amgen, outside the submitted manuscript. G.R. reports honoraria from Bristol-Myers-Squibb, Roche, Amgen, Jansenn, outside the submitted manuscript. A.P. reports personal fees from Roche, AstraZeneca and BMS, outside the submitted work. U.M. reports personal fees (for service in the speaker bureau and as advisor) from Boehringer Ingelheim, AstraZeneca, Roche, MSD, Amgen, Merck, Eli Lilly, Thermofisher, Diaceutics. All remaining authors have declared no conflict of interest.

References

  1. Denis, M.G.; Bennouna, J. Osimertinib for Front-Line Treatment of Locally Advanced or Metastatic EGFR-Mutant NSCLC Patients: Efficacy, Acquired Resistance and Perspectives for Subsequent Treatments. Cancer Manag. Res. 2020, 12, 12593–12602. [Google Scholar] [CrossRef]
  2. Tabbò, F.; Passiglia, F.; Novello, S. Upfront Management of ALK-Rearranged Metastatic Non-Small Cell Lung Cancer: One Inhibitor Fits All? Curr. Oncol. Rep. 2021, 23, 10. [Google Scholar] [CrossRef]
  3. Drilon, A.; Jenkins, C.; Iyer, S.; Schoenfeld, A.; Keddy, C.; Davare, M.A. ROS1-Dependent Cancers—Biology, Diagnostics and Therapeutics. Nat. Rev. Clin. Oncol. 2021, 18, 35–55. [Google Scholar] [CrossRef] [PubMed]
  4. Malapelle, U.; Rossi, G.; Pisapia, P.; Barberis, M.; Buttitta, F.; Castiglione, F.; Cecere, F.L.; Grimaldi, A.M.; Iaccarino, A.; Marchetti, A.; et al. BRAF as a Positive Predictive Biomarker: Focus on Lung Cancer and Melanoma Patients. Crit. Rev. Oncol. Hematol. 2020, 156, 103118. [Google Scholar] [CrossRef] [PubMed]
  5. Russo, A.; Lopes, A.R.; McCusker, M.G.; Garrigues, S.G.; Ricciardi, G.R.; Arensmeyer, K.E.; Scilla, K.A.; Mehra, R.; Rolfo, C. New Targets in Lung Cancer (Excluding EGFR, ALK, ROS1). Curr. Oncol. Rep. 2020, 22, 48. [Google Scholar] [CrossRef]
  6. Lamberti, G.; Andrini, E.; Sisi, M.; Rizzo, A.; Parisi, C.; Di Federico, A.; Gelsomino, F.; Ardizzoni, A. Beyond EGFR, ALK and ROS1: Current Evidence and Future Perspectives on Newly Targetable Oncogenic Drivers in Lung Adenocarcinoma. Crit. Rev. Oncol. Hematol. 2020, 156, 103119. [Google Scholar] [CrossRef] [PubMed]
  7. Suzawa, K.; Offin, M.; Schoenfeld, A.J.; Plodkowski, A.J.; Odintsov, I.; Lu, D.; Lockwood, W.W.; Arcila, M.E.; Rudin, C.M.; Drilon, A.; et al. Acquired MET Exon 14 Alteration Drives Secondary Resistance to Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor in EGFR-Mutated Lung Cancer. JCO Precis. Oncol. 2019, 3. [Google Scholar] [CrossRef]
  8. Dagogo-Jack, I.; Yoda, S.; Lennerz, J.K.; Langenbucher, A.; Lin, J.J.; Rooney, M.M.; Prutisto-Chang, K.; Oh, A.; Adams, N.A.; Yeap, B.Y.; et al. MET Alterations Are a Recurring and Actionable Resistance Mechanism in ALK-Positive Lung Cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2020, 26, 2535–2545. [Google Scholar] [CrossRef] [Green Version]
  9. Friedlaender, A.; Drilon, A.; Banna, G.L.; Peters, S.; Addeo, A. The METeoric Rise of MET in Lung Cancer. Cancer 2020, 126, 4826–4837. [Google Scholar] [CrossRef] [PubMed]
  10. Huang, C.; Zou, Q.; Liu, H.; Qiu, B.; Li, Q.; Lin, Y.; Liang, Y. Management of Non-Small Cell Lung Cancer Patients with MET Exon 14 Skipping Mutations. Curr. Treat. Options Oncol. 2020, 21, 33. [Google Scholar] [CrossRef]
  11. Salgia, R.; Sattler, M.; Scheele, J.; Stroh, C.; Felip, E. The Promise of Selective MET Inhibitors in Non-Small Cell Lung Cancer with MET Exon 14 Skipping. Cancer Treat. Rev. 2020, 87, 102022. [Google Scholar] [CrossRef]
  12. Awad, M.M.; Oxnard, G.R.; Jackman, D.M.; Savukoski, D.O.; Hall, D.; Shivdasani, P.; Heng, J.C.; Dahlberg, S.E.; Jänne, P.A.; Verma, S.; et al. MET Exon 14 Mutations in Non-Small-Cell Lung Cancer Are Associated With Advanced Age and Stage-Dependent MET Genomic Amplification and c-Met Overexpression. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2016, 34, 721–730. [Google Scholar] [CrossRef] [Green Version]
  13. Tong, J.H.; Yeung, S.F.; Chan, A.W.H.; Chung, L.Y.; Chau, S.L.; Lung, R.W.M.; Tong, C.Y.; Chow, C.; Tin, E.K.Y.; Yu, Y.H.; et al. MET Amplification and Exon 14 Splice Site Mutation Define Unique Molecular Subgroups of Non-Small Cell Lung Carcinoma with Poor Prognosis. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2016, 22, 3048–3056. [Google Scholar] [CrossRef] [Green Version]
  14. Lee, G.D.; Lee, S.E.; Oh, D.-Y.; Yu, D.-B.; Jeong, H.M.; Kim, J.; Hong, S.; Jung, H.S.; Oh, E.; Song, J.-Y.; et al. MET Exon 14 Skipping Mutations in Lung Adenocarcinoma: Clinicopathologic Implications and Prognostic Values. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2017, 12, 1233–1246. [Google Scholar] [CrossRef]
  15. Digumarthy, S.R.; Mendoza, D.P.; Zhang, E.W.; Lennerz, J.K.; Heist, R.S. Clinicopathologic and Imaging Features of Non-Small-Cell Lung Cancer with MET Exon 14 Skipping Mutations. Cancers 2019, 11, 2033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Bylicki, O.; Paleiron, N.; Assié, J.-B.; Chouaïd, C. Targeting the MET-Signaling Pathway in Non-Small-Cell Lung Cancer: Evidence to Date. OncoTargets Ther. 2020, 13, 5691–5706. [Google Scholar] [CrossRef]
  17. Liang, H.; Wang, M. MET Oncogene in Non-Small Cell Lung Cancer: Mechanism of MET Dysregulation and Agents Targeting the HGF/c-Met Axis. OncoTargets Ther. 2020, 13, 2491–2510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Yu, X.; Yu, S.; Fan, Y. Progress on Treatment of MET Signaling Pathway in Non-Small Cell Lung Cancer. Int. J. Clin. Oncol. 2020, 25, 1450–1458. [Google Scholar] [CrossRef]
  19. Drilon, A.; Camidge, D.R.; Ou, S.-H.I.; Clark, J.W.; Socinski, M.; Riely, G.J.; Winter, M.; Wang, S.; Monti, K.; Wilner, K.; et al. Efficacy and safety of crizotinib in patients (pts) with advanced MET exon 14-altered non-small cell lung cancer (NSCLC). Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2016, 34, 108. [Google Scholar] [CrossRef]
  20. Pfizer’s XALKORI® (Crizotinib) Receives FDA Breakthrough Therapy Designation in Two New Indications/Pfizer. Available online: https://www.pfizer.com/news/press-release/press-release-detail/pfizer_s_xalkori_crizotinib_receives_fda_breakthrough_therapy_designation_in_two_new_indications-0 (accessed on 23 January 2021).
  21. Drilon, A.; Clark, J.W.; Weiss, J.; Ou, S.-H.I.; Camidge, D.R.; Solomon, B.J.; Otterson, G.A.; Villaruz, L.C.; Riely, G.J.; Heist, R.S.; et al. Antitumor Activity of Crizotinib in Lung Cancers Harboring a MET Exon 14 Alteration. Nat. Med. 2020, 26, 47–51. [Google Scholar] [CrossRef]
  22. Landi, L.; Chiari, R.; Tiseo, M.; D’Incà, F.; Dazzi, C.; Chella, A.; Delmonte, A.; Bonanno, L.; Giannarelli, D.; Cortinovis, D.L.; et al. Crizotinib in MET-Deregulated or ROS1-Rearranged Pretreated Non-Small Cell Lung Cancer (METROS): A Phase II, Prospective, Multicenter, Two-Arms Trial. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019, 25, 7312–7319. [Google Scholar] [CrossRef] [Green Version]
  23. Moro-Sibilot, D.; Cozic, N.; Pérol, M.; Mazières, J.; Otto, J.; Souquet, P.J.; Bahleda, R.; Wislez, M.; Zalcman, G.; Guibert, S.D.; et al. Crizotinib in C-MET- or ROS1-Positive NSCLC: Results of the AcSé Phase II Trial. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2019, 30, 1985–1991. [Google Scholar] [CrossRef]
  24. Awad, M.M.; Leonardi, G.C.; Kravets, S.; Dahlberg, S.E.; Drilon, A.; Noonan, S.A.; Camidge, D.R.; Ou, S.-H.I.; Costa, D.B.; Gadgeel, S.M.; et al. Impact of MET Inhibitors on Survival among Patients with Non-Small Cell Lung Cancer Harboring MET Exon 14 Mutations: A Retrospective Analysis. Lung Cancer Amst. Neth. 2019, 133, 96–102. [Google Scholar] [CrossRef]
  25. National Comprehensive Cancer Network. Clinical Practice Guidelines in Oncology (NCCN Guidelines). Non-Small Cell Lung Cancer. Version 3.2021—February 19 2021. Available online: https://www.nccn.org/professionals/physician_gls/pdf/nscl_blocks.pdf (accessed on 24 February 2021).
  26. FDA Grants Accelerated Approval to Capmatinib for Metastatic Non-Small Cell Lung Cancer. (FDA website); FDA; 2020. Available online: https://www.fda.gov/drugs/drug-approvals-and-databases/fda-grants-accelerated-approval-capmatinib-metastatic-non-small-cell-lung-cancer#:~:text=On%20May%206%2C%202020%2C%20the,by%20an%20FDA%2Dapproved%20test (accessed on 15 November 2020).
  27. Wolf, J.; Seto, T.; Han, J.-Y.; Reguart, N.; Garon, E.B.; Groen, H.J.M.; Tan, D.S.W.; Hida, T.; de Jonge, M.; Orlov, S.V.; et al. Capmatinib in MET Exon 14-Mutated or MET-Amplified Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2020, 383, 944–957. [Google Scholar] [CrossRef] [PubMed]
  28. Paik, P.K.; Veillon, R.; Cortot, A.B.; Felip, E.; Sakai, H.; Mazieres, J.; Griesinger, F.; Horn, L.; Senellart, H.; Meerbeeck, J.P.V.; et al. Phase II Study of Tepotinib in NSCLC Patients with METex14 Mutations. J. Clin. Oncol. 2019, 37, 9005. [Google Scholar] [CrossRef]
  29. Paik, P.K.; Felip, E.; Veillon, R.; Sakai, H.; Cortot, A.B.; Garassino, M.C.; Mazieres, J.; Viteri, S.; Senellart, H.; Van Meerbeeck, J.; et al. Tepotinib in Non-Small-Cell Lung Cancer with MET Exon 14 Skipping Mutations. N. Engl. J. Med. 2020, 383, 931–943. [Google Scholar] [CrossRef]
  30. Tepotinib Breakthrough Therapy—News—Merck Global. Available online: https://www.merckgroup.com/en/news/tepotinib-breakthrough-therapy-designation-11-09-2019.html (accessed on 23 January 2021).
  31. Lu, S.; Fang, J.; Cao, L.; Li, X.; Guo, Q.; Zhou, J.; Cheng, Y.; Jiang, L.; Chen, Y.; Zhang, H.; et al. Abstract CT031: Preliminary Efficacy and Safety Results of Savolitinib Treating Patients with Pulmonary Sarcomatoid Carcinoma (PSC) and Other Types of Non-Small Cell Lung Cancer (NSCLC) Harboring MET Exon 14 Skipping Mutations. Cancer Res. 2019, 79, CT031. [Google Scholar] [CrossRef]
  32. De Mello, R.A.; Neves, N.M.; Amaral, G.A.; Lippo, E.G.; Castelo-Branco, P.; Pozza, D.H.; Tajima, C.C.; Antoniou, G. The Role of MET Inhibitor Therapies in the Treatment of Advanced Non-Small Cell Lung Cancer. J. Clin. Med. 2020, 9, 1918. [Google Scholar] [CrossRef] [PubMed]
  33. Yoshimura, K.; Inoue, Y.; Tsuchiya, K.; Karayama, M.; Yamada, H.; Iwashita, Y.; Kawase, A.; Tanahashi, M.; Ogawa, H.; Inui, N.; et al. Elucidation of the Relationships of MET Protein Expression and Gene Copy Number Status with PD-L1 Expression and the Immune Microenvironment in Non-Small Cell Lung Cancer. Lung Cancer Amst. Neth. 2020, 141, 21–31. [Google Scholar] [CrossRef]
  34. Mazieres, J.; Drilon, A.; Lusque, A.; Mhanna, L.; Cortot, A.B.; Mezquita, L.; Thai, A.A.; Mascaux, C.; Couraud, S.; Veillon, R.; et al. Immune Checkpoint Inhibitors for Patients with Advanced Lung Cancer and Oncogenic Driver Alterations: Results from the IMMUNOTARGET Registry. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2019, 30, 1321–1328. [Google Scholar] [CrossRef]
  35. Sabari, J.K.; Leonardi, G.C.; Shu, C.A.; Umeton, R.; Montecalvo, J.; Ni, A.; Chen, R.; Dienstag, J.; Mrad, C.; Bergagnini, I.; et al. PD-L1 Expression, Tumor Mutational Burden, and Response to Immunotherapy in Patients with MET Exon 14 Altered Lung Cancers. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2018, 29, 2085–2091. [Google Scholar] [CrossRef]
  36. Mayenga, M.; Assié, J.-B.; Monnet, I.; Massiani, M.-A.; Tabeze, L.; Friard, S.; Fraboulet, S.; Métivier, A.-C.; Chouaïd, C.; Zemoura, L.; et al. Durable Responses to Immunotherapy of Non-Small Cell Lung Cancers Harboring MET Exon-14-Skipping Mutation: A Series of 6 Cases. Lung Cancer Amst. Neth. 2020, 150, 21–25. [Google Scholar] [CrossRef]
  37. Guisier, F.; Dubos-Arvis, C.; Viñas, F.; Doubre, H.; Ricordel, C.; Ropert, S.; Janicot, H.; Bernardi, M.; Fournel, P.; Lamy, R.; et al. Efficacy and Safety of Anti-PD-1 Immunotherapy in Patients With Advanced NSCLC With BRAF, HER2, or MET Mutations or RET Translocation: GFPC 01-2018. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2020, 15, 628–636. [Google Scholar] [CrossRef]
  38. Platt, A.; Morten, J.; Ji, Q.; Elvin, P.; Womack, C.; Su, X.; Donald, E.; Gray, N.; Read, J.; Bigley, G.; et al. A Retrospective Analysis of RET Translocation, Gene Copy Number Gain and Expression in NSCLC Patients Treated with Vandetanib in Four Randomized Phase III Studies. BMC Cancer 2015, 15, 171. [Google Scholar] [CrossRef] [Green Version]
  39. Wang, R.; Hu, H.; Pan, Y.; Li, Y.; Ye, T.; Li, C.; Luo, X.; Wang, L.; Li, H.; Zhang, Y.; et al. RET Fusions Define a Unique Molecular and Clinicopathologic Subtype of Non-Small-Cell Lung Cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2012, 30, 4352–4359. [Google Scholar] [CrossRef]
  40. Michels, S.; Scheel, A.H.; Scheffler, M.; Schultheis, A.M.; Gautschi, O.; Aebersold, F.; Diebold, J.; Pall, G.; Rothschild, S.; Bubendorf, L.; et al. Clinicopathological Characteristics of RET Rearranged Lung Cancer in European Patients. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2016, 11, 122–127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Song, Z.; Yu, X.; Zhang, Y. Clinicopathologic Characteristics, Genetic Variability and Therapeutic Options of RET Rearrangements Patients in Lung Adenocarcinoma. Lung Cancer Amst. Neth. 2016, 101, 16–21. [Google Scholar] [CrossRef]
  42. Mahato, A.K.; Sidorova, Y.A. RET Receptor Tyrosine Kinase: Role in Neurodegeneration, Obesity, and Cancer. Int. J. Mol. Sci. 2020, 21, 7108. [Google Scholar] [CrossRef] [PubMed]
  43. Kotzbauer, P.T.; Lampe, P.A.; Heuckeroth, R.O.; Golden, J.P.; Creedon, D.J.; Johnson, E.M.; Milbrandt, J. Neurturin, a Relative of Glial-Cell-Line-Derived Neurotrophic Factor. Nature 1996, 384, 467–470. [Google Scholar] [CrossRef] [PubMed]
  44. Leitner, M.L.; Molliver, D.C.; Osborne, P.A.; Vejsada, R.; Golden, J.P.; Lampe, P.A.; Kato, A.C.; Milbrandt, J.; Johnson, E.M. Analysis of the Retrograde Transport of Glial Cell Line-Derived Neurotrophic Factor (GDNF), Neurturin, and Persephin Suggests That in Vivo Signaling for the GDNF Family Is GFRalpha Coreceptor-Specific. J. Neurosci. Off. J. Soc. Neurosci. 1999, 19, 9322–9331. [Google Scholar] [CrossRef] [Green Version]
  45. Baloh, R.H.; Tansey, M.G.; Lampe, P.A.; Fahrner, T.J.; Enomoto, H.; Simburger, K.S.; Leitner, M.L.; Araki, T.; Johnson, E.M.; Milbrandt, J. Artemin, a Novel Member of the GDNF Ligand Family, Supports Peripheral and Central Neurons and Signals through the GFRalpha3-RET Receptor Complex. Neuron 1998, 21, 1291–1302. [Google Scholar] [CrossRef] [Green Version]
  46. Qian, Y.; Chai, S.; Liang, Z.; Wang, Y.; Zhou, Y.; Xu, X.; Zhang, C.; Zhang, M.; Si, J.; Huang, F.; et al. KIF5B-RET Fusion Kinase Promotes Cell Growth by Multilevel Activation of STAT3 in Lung Cancer. Mol. Cancer 2014, 13, 176. [Google Scholar] [CrossRef] [PubMed]
  47. Pao, W.; Hutchinson, K.E. Chipping Away at the Lung Cancer Genome. Nat. Med. 2012, 18, 349–351. [Google Scholar] [CrossRef]
  48. Lipson, D.; Capelletti, M.; Yelensky, R.; Otto, G.; Parker, A.; Jarosz, M.; Curran, J.A.; Balasubramanian, S.; Bloom, T.; Brennan, K.W.; et al. Identification of New ALK and RET Gene Fusions from Colorectal and Lung Cancer Biopsies. Nat. Med. 2012, 18, 382–384. [Google Scholar] [CrossRef]
  49. Go, H.; Jung, Y.J.; Kang, H.W.; Park, I.-K.; Kang, C.-H.; Lee, J.W.; Ju, Y.S.; Seo, J.-S.; Chung, D.H.; Kim, Y.T. Diagnostic Method for the Detection of KIF5B-RET Transformation in Lung Adenocarcinoma. Lung Cancer Amst. Neth. 2013, 82, 44–50. [Google Scholar] [CrossRef] [PubMed]
  50. Mizukami, T.; Shiraishi, K.; Shimada, Y.; Ogiwara, H.; Tsuta, K.; Ichikawa, H.; Sakamoto, H.; Kato, M.; Shibata, T.; Nakano, T.; et al. Molecular Mechanisms Underlying Oncogenic RET Fusion in Lung Adenocarcinoma. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2014, 9, 622–630. [Google Scholar] [CrossRef] [Green Version]
  51. Tsuta, K.; Kohno, T.; Yoshida, A.; Shimada, Y.; Asamura, H.; Furuta, K.; Kushima, R. RET-Rearranged Non-Small-Cell Lung Carcinoma: A Clinicopathological and Molecular Analysis. Br. J. Cancer 2014, 110, 1571–1578. [Google Scholar] [CrossRef] [Green Version]
  52. Yoh, K.; Seto, T.; Satouchi, M.; Nishio, M.; Yamamoto, N.; Murakami, H.; Nogami, N.; Matsumoto, S.; Kohno, T.; Tsuta, K.; et al. Vandetanib in Patients with Previously Treated RET-Rearranged Advanced Non-Small-Cell Lung Cancer (LURET): An Open-Label, Multicentre Phase 2 Trial. Lancet Respir. Med. 2017, 5, 42–50. [Google Scholar] [CrossRef]
  53. Hida, T.; Velcheti, V.; Reckamp, K.L.; Nokihara, H.; Sachdev, P.; Kubota, T.; Nakada, T.; Dutcus, C.E.; Ren, M.; Tamura, T. A Phase 2 Study of Lenvatinib in Patients with RET Fusion-Positive Lung Adenocarcinoma. Lung Cancer Amst. Neth. 2019, 138, 124–130. [Google Scholar] [CrossRef] [Green Version]
  54. Horiike, A.; Takeuchi, K.; Uenami, T.; Kawano, Y.; Tanimoto, A.; Kaburaki, K.; Tambo, Y.; Kudo, K.; Yanagitani, N.; Ohyanagi, F.; et al. Sorafenib Treatment for Patients with RET Fusion-Positive Non-Small Cell Lung Cancer. Lung Cancer Amst. Neth. 2016, 93, 43–46. [Google Scholar] [CrossRef] [PubMed]
  55. Drilon, A.; Rekhtman, N.; Arcila, M.; Wang, L.; Ni, A.; Albano, M.; Van Voorthuysen, M.; Somwar, R.; Smith, R.S.; Montecalvo, J.; et al. Cabozantinib in Patients with Advanced RET-Rearranged Non-Small-Cell Lung Cancer: An Open-Label, Single-Centre, Phase 2, Single-Arm Trial. Lancet Oncol. 2016, 17, 1653–1660. [Google Scholar] [CrossRef] [Green Version]
  56. Drilon, A.; Oxnard, G.R.; Tan, D.S.W.; Loong, H.H.F.; Johnson, M.; Gainor, J.; McCoach, C.E.; Gautschi, O.; Besse, B.; Cho, B.C.; et al. Efficacy of Selpercatinib in RET Fusion-Positive Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2020, 383, 813–824. [Google Scholar] [CrossRef] [PubMed]
  57. ESMO FDA Approves Pralsetinib for Lung Cancer with RET Gene Fusions. Available online: https://www.esmo.org/oncology-news/fda-approves-pralsetinib-for-lung-cancer-with-ret-gene-fusions (accessed on 19 December 2020).
  58. Elfving, H.; Broström, E.; Moens, L.N.J.; Almlöf, J.; Cerjan, D.; Lauter, G.; Nord, H.; Mattsson, J.S.M.; Ullenhag, G.J.; Strell, C.; et al. Evaluation of NTRK Immunohistochemistry as a Screening Method for NTRK Gene Fusion Detection in Non-Small Cell Lung Cancer. Lung Cancer Amst. Neth. 2021, 151, 53–59. [Google Scholar] [CrossRef]
  59. Farago, A.F.; Taylor, M.S.; Doebele, R.C.; Zhu, V.W.; Kummar, S.; Spira, A.I.; Boyle, T.A.; Haura, E.B.; Arcila, M.E.; Benayed, R.; et al. Clinicopathologic Features of Non-Small-Cell Lung Cancer Harboring an NTRK Gene Fusion. JCO Precis. Oncol. 2018, 2018. [Google Scholar] [CrossRef]
  60. Ozono, K.; Ohishi, Y.; Onishi, H.; Nakamura, K.; Motoshita, J.; Kato, M.; Nakanishi, R.; Nakamura, M.; Oda, Y. Brain-Derived Neurotrophic Factor/Tropomyosin-Related Kinase B Signaling Pathway Contributes to the Aggressive Behavior of Lung Squamous Cell Carcinoma. Lab. Investig. J. Tech. Methods Pathol. 2017, 97, 1332–1342. [Google Scholar] [CrossRef]
  61. Okamura, K.; Harada, T.; Wang, S.; Ijichi, K.; Furuyama, K.; Koga, T.; Okamoto, T.; Takayama, K.; Yano, T.; Nakanishi, Y. Expression of TrkB and BDNF Is Associated with Poor Prognosis in Non-Small Cell Lung Cancer. Lung Cancer Amst. Neth. 2012, 78, 100–106. [Google Scholar] [CrossRef]
  62. Gatalica, Z.; Xiu, J.; Swensen, J.; Vranic, S. Molecular Characterization of Cancers with NTRK Gene Fusions. Mod. Pathol. Off. J. US Can. Acad. Pathol. Inc. 2019, 32, 147–153. [Google Scholar] [CrossRef]
  63. Haratake, N.; Seto, T. NTRK Fusion-Positive Non-Small-Cell Lung Cancer: The Diagnosis and Targeted Therapy. Clin. Lung Cancer 2021, 22, 1–5. [Google Scholar] [CrossRef]
  64. Liu, D.; Offin, M.; Harnicar, S.; Li, B.T.; Drilon, A. Entrectinib: An Orally Available, Selective Tyrosine Kinase Inhibitor for the Treatment of NTRK, ROS1, and ALK Fusion-Positive Solid Tumors. Ther. Clin. Risk Manag. 2018, 14, 1247–1252. [Google Scholar] [CrossRef] [Green Version]
  65. Kummar, S.; Lassen, U.N. TRK Inhibition: A New Tumor-Agnostic Treatment Strategy. Target. Oncol. 2018, 13, 545–556. [Google Scholar] [CrossRef] [Green Version]
  66. Marchiò, C.; Scaltriti, M.; Ladanyi, M.; Iafrate, A.J.; Bibeau, F.; Dietel, M.; Hechtman, J.F.; Troiani, T.; López-Rios, F.; Douillard, J.-Y.; et al. ESMO Recommendations on the Standard Methods to Detect NTRK Fusions in Daily Practice and Clinical Research. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2019, 30, 1417–1427. [Google Scholar] [CrossRef] [Green Version]
  67. Lee, J.; Park, S.; Jung, H.A.; Sun, J.-M.; Lee, S.-H.; Ahn, J.S.; Park, K.; Ahn, M.-J. Evaluating Entrectinib as a Treatment Option for Non-Small Cell Lung Cancer. Expert Opin. Pharmacother. 2020, 21, 1935–1942. [Google Scholar] [CrossRef] [PubMed]
  68. Sartore-Bianchi, A.; Pizzutilo, E.G.; Marrapese, G.; Tosi, F.; Cerea, G.; Siena, S. Entrectinib for the Treatment of Metastatic NSCLC: Safety and Efficacy. Expert Rev. Anticancer Ther. 2020, 20, 333–341. [Google Scholar] [CrossRef] [PubMed]
  69. Doebele, R.C.; Drilon, A.; Paz-Ares, L.; Siena, S.; Shaw, A.T.; Farago, A.F.; Blakely, C.M.; Seto, T.; Cho, B.C.; Tosi, D.; et al. Entrectinib in Patients with Advanced or Metastatic NTRK Fusion-Positive Solid Tumours: Integrated Analysis of Three Phase 1-2 Trials. Lancet Oncol. 2020, 21, 271–282. [Google Scholar] [CrossRef]
  70. Hong, D.S.; DuBois, S.G.; Kummar, S.; Farago, A.F.; Albert, C.M.; Rohrberg, K.S.; van Tilburg, C.M.; Nagasubramanian, R.; Berlin, J.D.; Federman, N.; et al. Larotrectinib in Patients with TRK Fusion-Positive Solid Tumours: A Pooled Analysis of Three Phase 1/2 Clinical Trials. Lancet Oncol. 2020, 21, 531–540. [Google Scholar] [CrossRef]
  71. Slebos, R.J.; Kibbelaar, R.E.; Dalesio, O.; Kooistra, A.; Stam, J.; Meijer, C.J.; Wagenaar, S.S.; Vanderschueren, R.G.; van Zandwijk, N.; Mooi, W.J. K-Ras Oncogene Activation as a Prognostic Marker in Adenocarcinoma of the Lung. N. Engl. J. Med. 1990, 323, 561–565. [Google Scholar] [CrossRef]
  72. Dearden, S.; Stevens, J.; Wu, Y.-L.; Blowers, D. Mutation Incidence and Coincidence in Non Small-Cell Lung Cancer: Meta-Analyses by Ethnicity and Histology (MutMap). Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2013, 24, 2371–2376. [Google Scholar] [CrossRef]
  73. Dogan, S.; Shen, R.; Ang, D.C.; Johnson, M.L.; D’Angelo, S.P.; Paik, P.K.; Brzostowski, E.B.; Riely, G.J.; Kris, M.G.; Zakowski, M.F.; et al. Molecular Epidemiology of EGFR and KRAS Mutations in 3,026 Lung Adenocarcinomas: Higher Susceptibility of Women to Smoking-Related KRAS-Mutant Cancers. Clin. Cancer Res. 2012, 18, 6169–6177. [Google Scholar] [CrossRef] [Green Version]
  74. Román, M.; Baraibar, I.; López, I.; Nadal, E.; Rolfo, C.; Vicent, S.; Gil-Bazo, I. KRAS Oncogene in Non-Small Cell Lung Cancer: Clinical Perspectives on the Treatment of an Old Target. Mol. Cancer 2018, 17, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Mitin, N.; Rossman, K.L.; Der, C.J. Signaling Interplay in Ras Superfamily Function. Curr. Biol. CB 2005, 15, 563–574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Shackelford, R.E.; Whitling, N.A.; McNab, P.; Japa, S.; Coppola, D. KRAS Testing: A Tool for the Implementation of Personalized Medicine. Genes Cancer 2012, 3, 459–466. [Google Scholar] [CrossRef] [Green Version]
  77. Torralvo, J.; Friedlaender, A.; Achard, V.; Addeo, A. The Activity of Immune Checkpoint Inhibition in KRAS Mutated Non-Small Cell Lung Cancer: A Single Centre Experience. Cancer Genom. Proteom. 2019, 16, 577–582. [Google Scholar] [CrossRef] [Green Version]
  78. Kim, J.H.; Kim, H.S.; Kim, B.J. Prognostic Value of KRAS Mutation in Advanced Non-Small-Cell Lung Cancer Treated with Immune Checkpoint Inhibitors: A Meta-Analysis and Review. Oncotarget 2017, 8, 48248–48252. [Google Scholar] [CrossRef] [Green Version]
  79. Dong, Z.-Y.; Zhong, W.-Z.; Zhang, X.-C.; Su, J.; Xie, Z.; Liu, S.-Y.; Tu, H.-Y.; Chen, H.-J.; Sun, Y.-L.; Zhou, Q.; et al. Potential Predictive Value of TP53 and KRAS Mutation Status for Response to PD-1 Blockade Immunotherapy in Lung Adenocarcinoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2017, 23, 3012–3024. [Google Scholar] [CrossRef] [Green Version]
  80. Skoulidis, F.; Goldberg, M.E.; Greenawalt, D.M.; Hellmann, M.D.; Awad, M.M.; Gainor, J.F.; Schrock, A.B.; Hartmaier, R.J.; Trabucco, S.E.; Gay, L.; et al. STK11/LKB1 Mutations and PD-1 Inhibitor Resistance in KRAS-Mutant Lung Adenocarcinoma. Cancer Discov. 2018, 8, 822–835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Hong, D.S.; Fakih, M.G.; Strickler, J.H.; Desai, J.; Durm, G.A.; Shapiro, G.I.; Falchook, G.S.; Price, T.J.; Sacher, A.; Denlinger, C.S.; et al. KRASG12C Inhibition with Sotorasib in Advanced Solid Tumors. N. Engl. J. Med. 2020, 383, 1207–1217. [Google Scholar] [CrossRef]
  82. Johnson, M.L.; Ou, S.H.I.; Barve, M.; Rybkin, I.I.; Papadopoulos, K.P.; Leal, T.A.; Velastegui, K.; Christensen, J.G.; Kheoh, T.; Chao, R.C.; et al. KRYSTAL-1: Activity and Safety of Adagrasib (MRTX849) in Patients with Colorectal Cancer (CRC) and Other Solid Tumors Harboring a KRAS G12C Mutation. Eur. J. Cancer 2020, 138, 2. [Google Scholar] [CrossRef]
  83. Puyol, M.; Martín, A.; Dubus, P.; Mulero, F.; Pizcueta, P.; Khan, G.; Guerra, C.; Santamaría, D.; Barbacid, M. A Synthetic Lethal Interaction between K-Ras Oncogenes and Cdk4 Unveils a Therapeutic Strategy for Non-Small Cell Lung Carcinoma. Cancer Cell 2010, 18, 63–73. [Google Scholar] [CrossRef]
  84. Goldman, J.W.; Mazieres, J.; Barlesi, F.; Dragnev, K.H.; Koczywas, M.; Göskel, T.; Cortot, A.B.; Girard, N.; Wesseler, C.; Bischoff, H.; et al. A Randomized Phase III Study of Abemaciclib Versus Erlotinib in Patients with Stage IV Non-Small Cell Lung Cancer With a Detectable KRAS Mutation Who Failed Prior Platinum-Based Therapy: JUNIPER. Front. Oncol. 2020, 10, 578756. [Google Scholar] [CrossRef]
  85. Samuels, Y. High Frequency of Mutations of the PIK3CA Gene in Human Cancers. Science 2004, 304, 554. [Google Scholar] [CrossRef] [Green Version]
  86. Scheffler, M.; Bos, M.; Gardizi, M.; König, K.; Michels, S.; Fassunke, J.; Heydt, C.; Künstlinger, H.; Ihle, M.; Ueckeroth, F.; et al. PIK3CA Mutations in Non-Small Cell Lung Cancer (NSCLC): Genetic Heterogeneity, Prognostic Impact and Incidence of Prior Malignancies. Oncotarget 2015, 6, 1315–1326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Wang, L.; Hu, H.; Pan, Y.; Wang, R.; Li, Y.; Shen, L.; Yu, Y.; Li, H.; Cai, D.; Sun, Y.; et al. PIK3CA Mutations Frequently Coexist with EGFR/KRAS Mutations in Non-Small Cell Lung Cancer and Suggest Poor Prognosis in EGFR/KRAS Wildtype Subgroup. PLoS ONE 2014, 9, e88291. [Google Scholar] [CrossRef] [Green Version]
  88. Wang, Y.; Wang, Y.; Li, J.; Li, J.; Che, G. Clinical Significance of PIK3CA Gene in Non-Small-Cell Lung Cancer: A Systematic Review and Meta-Analysis. BioMed Res. Int. 2020, 2020, 1–9. [Google Scholar] [CrossRef]
  89. Bar, J.; Damianovich, M.; Hout Siloni, G.; Dar, E.; Cohen, Y.; Perelman, M.; Ben Nun, A.; Simansky, D.; Yellin, A.; Urban, D.; et al. Genetic Mutation Screen in Early Non–Small-Cell Lung Cancer (NSCLC) Specimens. Clin. Lung Cancer 2014, 15, 159–165. [Google Scholar] [CrossRef] [PubMed]
  90. Alqahtani, A.; Ayesh, H.S.K.; Halawani, H. PIK3CA Gene Mutations in Solid Malignancies: Association with Clinicopathological Parameters and Prognosis. Cancers 2019, 12, 93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Sarris, E.G.; Saif, M.W.; Syrigos, K.N. The Biological Role of PI3K Pathway in Lung Cancer. Pharm. Basel Switz. 2012, 5, 1236–1264. [Google Scholar] [CrossRef] [Green Version]
  92. Cheng, H.; Shcherba, M.; Pendurti, G.; Liang, Y.; Piperdi, B.; Perez-Soler, R. Targeting the PI3K/AKT/MTOR Pathway: Potential for Lung Cancer Treatment. Lung Cancer Manag. 2014, 3, 67–75. [Google Scholar] [CrossRef] [Green Version]
  93. Engelman, J.A.; Luo, J.; Cantley, L.C. The Evolution of Phosphatidylinositol 3-Kinases as Regulators of Growth and Metabolism. Nat. Rev. Genet. 2006, 7, 606–619. [Google Scholar] [CrossRef]
  94. Vansteenkiste, J.F.; Canon, J.-L.; De Braud, F.; Grossi, F.; De Pas, T.; Gray, J.E.; Su, W.-C.; Felip, E.; Yoshioka, H.; Gridelli, C.; et al. Safety and Efficacy of Buparlisib (BKM120) in Patients with PI3K Pathway-Activated Non-Small Cell Lung Cancer: Results from the Phase II BASALT-1 Study. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2015, 10, 1319–1327. [Google Scholar] [CrossRef] [Green Version]
  95. Yang, J.; Nie, J.; Ma, X.; Wei, Y.; Peng, Y.; Wei, X. Targeting PI3K in Cancer: Mechanisms and Advances in Clinical Trials. Mol. Cancer 2019, 18, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Tan, A.C. Targeting the PI3K/Akt/MTOR Pathway in Non-Small Cell Lung Cancer (NSCLC). Thorac. Cancer 2020, 11, 511–518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Schram, A.M.; Gandhi, L.; Mita, M.M.; Damstrup, L.; Campana, F.; Hidalgo, M.; Grande, E.; Hyman, D.M.; Heist, R.S. A Phase Ib Dose-Escalation and Expansion Study of the Oral MEK Inhibitor Pimasertib and PI3K/MTOR Inhibitor Voxtalisib in Patients with Advanced Solid Tumours. Br. J. Cancer 2018, 119, 1471–1476. [Google Scholar] [CrossRef]
  98. Jänne, P.A.; Cohen, R.B.; Laird, A.D.; Macé, S.; Engelman, J.A.; Ruiz-Soto, R.; Rockich, K.; Xu, J.; Shapiro, G.I.; Martinez, P.; et al. Phase I Safety and Pharmacokinetic Study of the PI3K/MTOR Inhibitor SAR245409 (XL765) in Combination with Erlotinib in Patients with Advanced Solid Tumors. J. Thorac. Oncol. 2014, 9, 316–323. [Google Scholar] [CrossRef] [Green Version]
  99. Soria, J.-C.; Adjei, A.A.; Bahleda, R.; Besse, B.; Ferte, C.; Planchard, D.; Zhou, J.; Ware, J.; Morrissey, K.; Shankar, G.; et al. A Phase IB Dose-Escalation Study of the Safety and Pharmacokinetics of Pictilisib in Combination with Either Paclitaxel and Carboplatin (with or without Bevacizumab) or Pemetrexed and Cisplatin (with or without Bevacizumab) in Patients with Advanced Non-Small Cell Lung Cancer. Eur. J. Cancer Oxf. Engl. 1990 2017, 86, 186–196. [Google Scholar] [CrossRef]
  100. Levy, B.; Spira, A.; Becker, D.; Evans, T.; Schnadig, I.; Camidge, D.R.; Bauman, J.E.; Hausman, D.; Walker, L.; Nemunaitis, J.; et al. A Randomized, Phase 2 Trial of Docetaxel with or without PX-866, an Irreversible Oral Phosphatidylinositol 3-Kinase Inhibitor, in Patients with Relapsed or Metastatic Non-Small-Cell Lung Cancer. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2014, 9, 1031–1035. [Google Scholar] [CrossRef] [Green Version]
  101. Shapiro, G.I.; Rodon, J.; Bedell, C.; Kwak, E.L.; Baselga, J.; Braña, I.; Pandya, S.S.; Scheffold, C.; Laird, A.D.; Nguyen, L.T.; et al. Phase I Safety, Pharmacokinetic, and Pharmacodynamic Study of SAR245408 (XL147), an Oral Pan-Class I PI3K Inhibitor, in Patients with Advanced Solid Tumors. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2014, 20, 233–245. [Google Scholar] [CrossRef] [Green Version]
  102. Soria, J.-C.; LoRusso, P.; Bahleda, R.; Lager, J.; Liu, L.; Jiang, J.; Martini, J.-F.; Macé, S.; Burris, H. Phase I Dose-Escalation Study of Pilaralisib (SAR245408, XL147), a Pan-Class I PI3K Inhibitor, in Combination with Erlotinib in Patients with Solid Tumors. The Oncologist 2015, 20, 245–246. [Google Scholar] [CrossRef] [Green Version]
  103. Calvo, E.; Boni, V.; Heist, R.S.; Matulonis, U.; Janne, P.A.; Hamid, O.; Holgado, E.; Ordoñez, J.M.; Nunez, J.; Ugrenovic, M.; et al. 1230PD—Phase 1B Study of Oral Dual-Pi3K/Mtor Inhibitor Gdc-0980 in Combination with Carboplatin (Carbo)/Paclitaxel (Pac) ± Bevacizumab (Bev) and Cisplatin (Cis)/Pemetrexed (Pem) in Patients (Pts) with Advanced Solid Tumors and Nsclc. Ann. Oncol. 2014, 25, iv429. [Google Scholar] [CrossRef] [Green Version]
  104. Langer, C.J.; Redman, M.W.; Wade, J.L.; Aggarwal, C.; Bradley, J.D.; Crawford, J.; Stella, P.J.; Knapp, M.H.; Miao, J.; Minichiello, K.; et al. SWOG S1400B (NCT02785913), a Phase II Study of GDC-0032 (Taselisib) for Previously Treated PI3K-Positive Patients with Stage IV Squamous Cell Lung Cancer (Lung-MAP Sub-Study). J. Thorac. Oncol. 2019, 14, 1839–1846. [Google Scholar] [CrossRef]
  105. Arcila, M.E.; Chaft, J.E.; Nafa, K.; Roy-Chowdhuri, S.; Lau, C.; Zaidinski, M.; Paik, P.K.; Zakowski, M.F.; Kris, M.G.; Ladanyi, M. Prevalence, Clinicopathologic Associations, and Molecular Spectrum of ERBB2 (HER2) Tyrosine Kinase Mutations in Lung Adenocarcinomas. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2012, 18, 4910–4918. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Suzuki, M.; Shiraishi, K.; Yoshida, A.; Shimada, Y.; Suzuki, K.; Asamura, H.; Furuta, K.; Kohno, T.; Tsuta, K. HER2 Gene Mutations in Non-Small Cell Lung Carcinomas: Concurrence with Her2 Gene Amplification and Her2 Protein Expression and Phosphorylation. Lung Cancer Amst. Neth. 2015, 87, 14–22. [Google Scholar] [CrossRef] [PubMed]
  107. Robichaux, J.P.; Elamin, Y.Y.; Vijayan, R.S.K.; Nilsson, M.B.; Hu, L.; He, J.; Zhang, F.; Pisegna, M.; Poteete, A.; Sun, H.; et al. Pan-Cancer Landscape and Analysis of ERBB2 Mutations Identifies Poziotinib as a Clinically Active Inhibitor and Enhancer of T-DM1 Activity. Cancer Cell 2020, 37, 420. [Google Scholar] [CrossRef] [PubMed]
  108. Kris, M.G.; Johnson, B.E.; Berry, L.D.; Kwiatkowski, D.J.; Iafrate, A.J.; Wistuba, I.I.; Varella-Garcia, M.; Franklin, W.A.; Aronson, S.L.; Su, P.-F.; et al. Using Multiplexed Assays of Oncogenic Drivers in Lung Cancers to Select Targeted Drugs. JAMA 2014, 311, 1998–2006. [Google Scholar] [CrossRef]
  109. Li, B.T.; Ross, D.S.; Aisner, D.L.; Chaft, J.E.; Hsu, M.; Kako, S.L.; Kris, M.G.; Varella-Garcia, M.; Arcila, M.E. HER2 Amplification and HER2 Mutation Are Distinct Molecular Targets in Lung Cancers. J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2016, 11, 414–419. [Google Scholar] [CrossRef] [Green Version]
  110. Cancer Genome Atlas Research Network Comprehensive Molecular Profiling of Lung Adenocarcinoma. Nature 2014, 511, 543–550. [CrossRef]
  111. Arcila, M.E.; Nafa, K.; Chaft, J.E.; Rekhtman, N.; Lau, C.; Reva, B.A.; Zakowski, M.F.; Kris, M.G.; Ladanyi, M. EGFR Exon 20 Insertion Mutations in Lung Adenocarcinomas: Prevalence, Molecular Heterogeneity, and Clinicopathologic Characteristics. Mol. Cancer Ther. 2013, 12, 220–229. [Google Scholar] [CrossRef] [Green Version]
  112. Lv, Q.; Meng, Z.; Yu, Y.; Jiang, F.; Guan, D.; Liang, C.; Zhou, J.; Lu, A.; Zhang, G. Molecular Mechanisms and Translational Therapies for Human Epidermal Receptor 2 Positive Breast Cancer. Int. J. Mol. Sci. 2016, 17, 2095. [Google Scholar] [CrossRef] [PubMed]
  113. Wei, X.-W.; Gao, X.; Zhang, X.-C.; Yang, J.-J.; Chen, Z.-H.; Wu, Y.-L.; Zhou, Q. Mutational Landscape and Characteristics of ERBB2 in Non-Small Cell Lung Cancer. Thorac. Cancer 2020, 11, 1512–1521. [Google Scholar] [CrossRef] [Green Version]
  114. Ross, D.S.; Zehir, A.; Cheng, D.T.; Benayed, R.; Nafa, K.; Hechtman, J.F.; Janjigian, Y.Y.; Weigelt, B.; Razavi, P.; Hyman, D.M.; et al. Next-Generation Assessment of Human Epidermal Growth Factor Receptor 2 (ERBB2) Amplification Status: Clinical Validation in the Context of a Hybrid Capture-Based, Comprehensive Solid Tumor Genomic Profiling Assay. J. Mol. Diagn. 2017, 19, 244–254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Budiarto, B.R. Desriani Dataset Reporting Detection of Breast Cancer-Related HER2I655V Polymorphism Using Allele-Specific Polymerase Chain Reaction. Data Brief 2016, 9, 689–695. [Google Scholar] [CrossRef] [Green Version]
  116. Imyanitov, E.N.; Iyevleva, A.G.; Levchenko, E.V. Molecular Testing and Targeted Therapy for Non-Small Cell Lung Cancer: Current Status and Perspectives. Crit. Rev. Oncol. Hematol. 2021, 157, 103194. [Google Scholar] [CrossRef] [PubMed]
  117. Van de Vijver, M. Emerging Technologies for HER2 Testing. Oncology 2002, 63 (Suppl. 1), 33–38. [Google Scholar] [CrossRef]
  118. Schaller, G.; Evers, K.; Papadopoulos, S.; Ebert, A.; Bühler, H. Current Use of HER2 Tests. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2001, 12, 97–100. [Google Scholar] [CrossRef]
  119. Jing, C.; Mao, X.; Wang, Z.; Sun, K.; Ma, R.; Wu, J.; Cao, H. Next-generation Sequencing-based Detection of EGFR, KRAS, BRAF, NRAS, PIK3CA, Her-2 and TP53 Mutations in Patients with Non-small Cell Lung Cancer. Mol. Med. Rep. 2018, 18, 2191–2197. [Google Scholar] [CrossRef] [PubMed]
  120. Smit, E.F.; Nakagawa, K.; Nagasaka, M.; Felip, E.; Goto, Y.; Li, B.T.; Murakami, H.; Barlesi, F.; Saltos, A.; Perol, M.; et al. Trastuzumab Deruxtecan (T-DXd; DS-8201) in Patients With HER2-Mutated Metastatic Non-Small Cell Lung Cancer: Interim Results of DESTINY-Lung0. 5. J. Clin. Oncol. 2020, 38, 9504. [Google Scholar] [CrossRef]
  121. Shitara, K.; Bang, Y.-J.; Iwasa, S.; Sugimoto, N.; Ryu, M.-H.; Sakai, D.; Chung, H.-C.; Kawakami, H.; Yabusaki, H.; Lee, J.; et al. Trastuzumab Deruxtecan in Previously Treated HER2-Positive Gastric Cancer. N. Engl. J. Med. 2020, 382, 2419–2430. [Google Scholar] [CrossRef]
  122. Socinski, M.A.; Cornelissen, R.; Garassino, M.C.; Clarke, J.; Tchekmedyian, N.; Molina, J.; Goldman, J.W.; Bhat, G.; Lebel, F.; Le, X. LBA60 ZENITH20, a Multinational, Multi-Cohort Phase II Study of Poziotinib in NSCLC Patients with EGFR or HER2 Exon 20 Insertion Mutations. Ann. Oncol. 2020, 31, 1188. [Google Scholar] [CrossRef]
  123. Prelaj, A.; Bottiglieri, A.; Proto, C.; Lo Russo, G.; Signorelli, D.; Ferrara, R.; Galli, G.; Toma, A.; Viscardi, G.; Brambilla, M.; et al. 1388P Poziotinib in Advanced NSCLC with EGFR or HER2 Exon 20 Insertion Mutation: Initial Results from a Single Site Expanded Access Program. Ann. Oncol. 2020, 31, 882. [Google Scholar] [CrossRef]
  124. Wang, Y.; Jiang, T.; Qin, Z.; Jiang, J.; Wang, Q.; Yang, S.; Rivard, C.; Gao, G.; Ng, T.L.; Tu, M.M.; et al. HER2 Exon 20 Insertions in Non-Small-Cell Lung Cancer Are Sensitive to the Irreversible Pan-HER Receptor Tyrosine Kinase Inhibitor Pyrotinib. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2019, 30, 447–455. [Google Scholar] [CrossRef] [Green Version]
  125. Gao, G.; Li, X.; Wang, Q.; Zhang, Y.; Chen, J.; Shu, Y.; Hu, Y.; Fan, Y.; Fang, J.; Chen, G.; et al. Single-Arm, Phase II Study of Pyrotinib in Advanced Non-Small Cell Lung Cancer (NSCLC) Patients with HER2 Exon 20 Mutation. J. Clin. Oncol. 2019, 37, 9089. [Google Scholar] [CrossRef]
  126. Janne, P.A.; Neal, J.W.; Camidge, D.R.; Spira, A.I.; Piotrowska, Z.; Horn, L.; Costa, D.B.; Tsao, A.S.; Patel, J.D.; Gadgeel, S.M.; et al. Antitumor Activity of TAK-788 in NSCLC with EGFR Exon 20 Insertions. J. Clin. Oncol. 2019, 37, 9007. [Google Scholar] [CrossRef]
  127. Dziadziuszko, R.; Smit, E.F.; Dafni, U.; Wolf, J.; Wasąg, B.; Biernat, W.; Finn, S.P.; Kammler, R.; Tsourti, Z.; Rabaglio, M.; et al. Afatinib in NSCLC With HER2 Mutations: Results of the Prospective, Open-Label Phase II NICHE Trial of European Thoracic Oncology Platform (ETOP). J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer 2019, 14, 1086–1094. [Google Scholar] [CrossRef]
  128. Hotta, K.; Aoe, K.; Kozuki, T.; Ohashi, K.; Ninomiya, K.; Ichihara, E.; Kubo, T.; Ninomiya, T.; Chikamori, K.; Harada, D.; et al. A Phase II Study of Trastuzumab Emtansine in HER2-Positive Non-Small-Cell Lung Cancer. J. Thorac. Oncol. 2017, 13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Kinoshita, I.; Goda, T.; Watanabe, K.; Maemondo, M.; Oizumi, S.; Amano, T.; Hatanaka, Y.; Matsuno, Y.; Nishihara, H.; Asahina, H.; et al. A Phase II Study of Trastuzumab Monotherapy in Pretreated Patients with Non-Small Cell Lung Cancers (NSCLCs) Harboring HER2 Alterations: HOT1303-B Trial. Ann Oncol 2018, 29, 540. [Google Scholar] [CrossRef]
  130. Gainor, J.F.; Dardaei, L.; Yoda, S.; Friboulet, L.; Leshchiner, I.; Katayama, R.; Dagogo-Jack, I.; Gadgeel, S.; Schultz, K.; Singh, M.; et al. Molecular Mechanisms of Resistance to First- and Second-Generation ALK Inhibitors in ALK-Rearranged Lung Cancer. Cancer Discov. 2016, 6, 1118–1133. [Google Scholar] [CrossRef] [Green Version]
  131. Van der Velden, D.L.; van Herpen, C.M.L.; van Laarhoven, H.W.M.; Smit, E.F.; Groen, H.J.M.; Willems, S.M.; Nederlof, P.M.; Langenberg, M.H.G.; Cuppen, E.; Sleijfer, S.; et al. Molecular Tumor Boards: Current Practice and Future Needs. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2017, 28, 3070–3075. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Molecular pathways of novel emerging targets in NSCLC and agents in clinical development. Several molecular pathways are physiologically activated by the interactions between circulating growth factors (colored circles) and trans-membrane receptors (colored sticks crossing the cell membrane), which result in the downstream activation of intracellular proteins (colored ovoids) associated with cell proliferation, increased aggressiveness, or immune escape. The details for each individual pathway are described in the appropriate paragraphs. Molecular alterations (e.g., gene fusions) potentially associated with tumorigenesis are reported next to the relevant molecule. When available, investigational agents able to inhibit specific pathways are reported (blank boxes), with reference to the specifically targeted molecule or interaction.
Figure 1. Molecular pathways of novel emerging targets in NSCLC and agents in clinical development. Several molecular pathways are physiologically activated by the interactions between circulating growth factors (colored circles) and trans-membrane receptors (colored sticks crossing the cell membrane), which result in the downstream activation of intracellular proteins (colored ovoids) associated with cell proliferation, increased aggressiveness, or immune escape. The details for each individual pathway are described in the appropriate paragraphs. Molecular alterations (e.g., gene fusions) potentially associated with tumorigenesis are reported next to the relevant molecule. When available, investigational agents able to inhibit specific pathways are reported (blank boxes), with reference to the specifically targeted molecule or interaction.
Ijms 22 02625 g001
Table 1. Principal single-arm Phase I–II trials on MET-TKI in METex14-mutated advanced NSCLC patients.
Table 1. Principal single-arm Phase I–II trials on MET-TKI in METex14-mutated advanced NSCLC patients.
DrugTrialPhaseTherapy Line *N ptsMain ResultsStatus
Multikinase inhibitors
CrizotinibPROFILE-1001
(NCT00585195)
I1L
≥2L
Tot 69
26 in 1L
43 in ≥2L
ORR tot = 32%
ORR 1L = 25%
ORR 2L = 37%
mDOR = 9.1 mo
mPFS = 7.3 mo
mOS = 20.5 mo
Closed
FDA-Approved
AcSé trial
(NCT02034981)
II≥1L25ORR = 40%
mDOR = 2.4 mo
mPFS = 3.6 mo
mOS = 9.5 mo
Closed
METROS
(NCT02499614)
II≥2L9ORR = 20%
mDOR = NE
mPFS = 2.6 mo
mOS = 3.8 mo
Closed
MET-specific TKI
CapmatinibGEOMETRY-mono-1
(NCT02414139)
II1L
2L,3L
28 in 1LORR = 68%
mDOR = 12.6 mo
mPFS = 12.4 mo
Ongoing
FDA-Approved
69 in 2L,3LORR = 41%
mDOR = 9.7 mo
mPFS = 5.4 mo
TepotinibVISION
(NCT02864992)
II1L,2L,3L66
diagnosed with LBx
ORR = 48%
mDOR = 9.9 mo
mPFS = 8.5 mo
Ongoing
(preliminary data)
FDA breakthrough designation
60
diagnosed with TBx
ORR = 50%
mDOR = 15.7 mo
mPFS = 11.0 mo
99
diagnosed with LBx + TBx
ORR = 46%
mDOR = 11.1 mo
mPFS = 8.5 mo
SavolitinibNCT02897479II≥2L70ORR = 47.5%
mDOR = NR
mPFS = 6.8 mo
Ongoing
MET—mesenchymal-epithelial transition factor; TKI—tyrosine kinase inhibitor; 1L—first-line; 2L—second-line; 3L—third-line; N—number; pts—patients; LBx—liquid biopsy; TBx—tissue biopsy; ORR—overall response rate; mo—month; mDOR—median duration of response; NE—not estimable; mPFS—median progression-free survival; mOS—median overall survival; NR—not reached; FDA—Food and Drug Administration. An asterisk (*) indicates the line or lines of treatment for advanced NSCLC in which the investigational agent or regimen was employed in each reported trial.
Table 2. Principal Phase Ib–II trials on MET-TKI in MET-amplified EGFR-mutated advanced NSCLC patients.
Table 2. Principal Phase Ib–II trials on MET-TKI in MET-amplified EGFR-mutated advanced NSCLC patients.
DrugTrialPhaseTherapy Line *Prior TreatmentTreatment ArmN ptsMain ResultsStatus
Single agent
CapmatinibGEOMETRY
-mono-1
(NCT02414139)
II≥2LNACapmatinib69ORR = 29%
mDOR = 8.3 mo
mPFS = 4.1 mo
Ongoing
Combinations
TepotinibINSIGH
(NCT01982955)
Ib/II2L1G-2G
EGFR-TKIs
Tepotinib + Gefitinib
vs.
Platinum-based CT
19ORR = 67% vs. 43%
mPFS = 16.6 vs. 4.2 mo
mOS = 37.3 vs. 13.1 mo
Closed
CapmatinibNCT01610336II2LGefitinib/
Erlotinib
Capmatinib
+ Gefitinib
100ORR = 47%Closed
CabozantinibNCI 9303 II (NCT01866410)II≥2L1G, 2G, 3G
EGFR-TKIs
Cabozantinib
+ Erlotinib
37ORR = 10.8%
mPFS = 3.6 mo
mOS = 13.1 mo
Closed
2L—second-line; NA—not available; 1G—first-generation; 2G—second-generation; EGFR—epidermal growth factor receptor; TKI—tyrosine kinase inhibitor; CT—chemotherapy; N—number; pts—patients; ORR—overall response rate; mo—month; mDOR—median duration of response; mPFS—median progression-free survival; vs.—versus; mOS—median overall survival. An asterisk (*) indicates the line or lines of treatment for advanced NSCLC in which the investigational agent or regimen was employed in each reported trial.
Table 3. Principal single-arm Phase I–II trials on TKI in RET-rearranged advanced NSCLC patients.
Table 3. Principal single-arm Phase I–II trials on TKI in RET-rearranged advanced NSCLC patients.
DrugTrialPhaseTherapy Line *N ptsMain ResultsStatus
SelpercatinibLIBRETTO-001
(NCT03157128)
II≥1L39 in 1LORR = 85%
1y-PFS = 75%
Closed
FDA approved
105 in ≥2L ORR = 64%
mDOR = 17.5 mo
mPFS = 16.5 mo
1yPFS = 66%
PralsetinibARROW
(NCT03037385)
I/II≥1L79ORR = 56%Ongoing
FDA approved
VandetanibLURET
(UMIN000010095 **)
II≥2L19ORR = 53%Ongoing
(preliminary data)
LenvatinibNCT01877083II≥1L25ORR = 16%Ongoing
(preliminary data)
Sorafenib000007515 **II≥2L3ORR = 0%Closed
CabozantinibNCT01639508II≥1L26ORR = 28%
mDOR = 7 mo
mPFS = 5.5 mo
mOS = 9.9 mo
Closed
1L—first-line; 2L—second-line; N—number; pts—patients; ORR—overall response rate; 1y-PFS—progression-free survival at one year; mo—month; mDOR—median duration of response; mPFS—median progression-free survival; mOS—median overall survival; FDA—Food and Drug Administration. An asterisk (*) indicates the line or lines of treatment for advanced NSCLC in which the investigational agent or regimen was employed in each reported trial. Two asterisks (**) indicate the UMIN trial number.
Table 4. Principal single-arm Phase I–II trials on NTRK inhibitors in NTRK-fused advanced NSCLC patients.
Table 4. Principal single-arm Phase I–II trials on NTRK inhibitors in NTRK-fused advanced NSCLC patients.
DrugTrialPhaseTherapy Line *N ptsMain ResultsStatus ***
EntrectinibALKA-372-001
(EudraCT2012–000148–88)
IAny10 **ORR = 70%
mDOR = 12.9 mo
mPFS = 14.9 mo
mOS = 23.9 mo
Closed
STARTRK-1
(NCT02097810)
IAnyClosed
STARTRK-2
(NCT02568267)
IIAnyOngoing
LarotrectinibLOXO-TRK-14001
(NCT02122913)
IAny12 **ORR = 75%
mDOR = NE
mPFS = 28.3 mo
mOS = 44.4 mo
Closed
LOXO-TRK-15003
(NCT02637687)
I/IIAnyOngoing
NAVIGATE
(NCT02576431)
IIAnyOngoing
N—number; pts patients; ORR—overall response rate; mo—month; mDOR—median duration of response; mPFS—median progression-free survival; mOS—median overall survival; NE—not estimable. An asterisk (*) indicates the line or lines of treatment for advanced NSCLC in which the investigational agent or regimen was employed in each reported trial. Two asterisks (**) indicate that the studies were pooled analyses. Three asterisks (***) indicate that Entrectinib and Larotrectinib FDA approved for NTRK-fused solid tumors.
Table 5. Principal Phase Ib–III trials on TKI in KRAS-mutated advanced NSCLC patients.
Table 5. Principal Phase Ib–III trials on TKI in KRAS-mutated advanced NSCLC patients.
DrugTrialPhaseTherapy Line *Treatment ArmN ptsMain ResultsStatus
Targeting KRAS pathway
Selumetinib
(MAPK inhibitor)
SELECT-1
(NCT01933932)
III2LSelumetinib + Docetaxel
vs.
Placebo + Docetaxel
510mPFS = 3.9 vs. 2.8 mo
mOS = 8.7 vs. 7.9 mo
ORR = 20.1% vs. 13.7%
mDOR = 2.9 vs. 4.5 mo
Closed
IND.219
(NCT02337530)
II1LARM A:
Salumetinib intermittent + Pemetrexed/Platinum
20ORR = 35%
mPFS = 7.5 mo
Closed
ARM B:
Salumetinib continuous + Pemetrexed/Platinum
21ORR = 62%
mPFS = 6.7 mo
ARM C:
Pemetrexed/Platinum
21ORR = 24%
mPFS = 4.0 mo
Trametinib
(MEK 1-2 inhibitor)
NCT01362296II≥2LTrametinib
vs.
Docetaxel
134mPFS = 12 vs. 11 weeks
mOS = 8 mo vs. NR
ORR = 12% vs. 12%
Closed
Momelotinib
(JAK1-2 inhibitor)
NCT02258607Ib≥2LMomelotinib
+ Trametinib
21ORR = NR
DCR = 57.1%
mPFS = 3.6 mo
mOS = 7.4 mo
Closed
Defactinib
(FAK inhibitor)
NCT01951690II≥2LDefactinib5512wks-PFS = 28%Closed
Targeting KRAS G12C
Sotorasib (AMG510)CODEBREAK 100
(NCT03600883)
I/II≥2LSotorasib129
(59 NSCLC)
ORR = 32.2%
DCR = 88.1%
mPFS = 6.3 mo
Ongoing
Adagrasib
(MRTX849)
KRYSTAL-1
(NCT03785249)
I/II≥1LAdagrasib110
(79 NSCLC)
ORR = 45%
DCR = 96%
Ongoing
Targeting CDK 4/6
Abemaciclib
(LY2835219)
JUNIPER
(NCT02152631)
III≥2LAbemaciclib
vs
Erlotinib
453mOS = 7.4 vs. 7.8 mo
mPFS = 3.6 vs. 1.9 mo
ORR = 8.9% vs. 2.7%
DCR = 54.5% vs. 31.7%
Ongoing
1L—first-line; 2L—second-line; vs.—versus; N—number; pts—patients; mPFS—median progression-free survival; mOS—median overall survival; mo—month; ORR—overall response rate; DCR—disease control rate; mDOR—median duration of response; 12wks-PFS—progression-free survival at 12 weeks; NR—not reached. An asterisk (*) indicates the line or lines of treatment for advanced NSCLC in which the investigational agent or regimen was employed in each reported trial.
Table 6. Principal Phase I–II trials on PI3K or PI3K/mTOR inhibitors in PI3K-mutated advanced NSCLC patients.
Table 6. Principal Phase I–II trials on PI3K or PI3K/mTOR inhibitors in PI3K-mutated advanced NSCLC patients.
DrugTrialPhaseTherapy LineTreatment Arms ***N ptsMain ResultsStatus
Selective PI3K inhibitor (anti p110α)
GDC-0032 (Taselisib)Lung-MAP
sub-study (NCT02785913)
II≥2L *Taselisib26 SqORR = 4.8%
mPFS = 2.9 mo
mOS = 5.9 mo
Closed
at futility
analysis
Pan-PI3K inhibitor
BKM120
(Buparlisib)
BASALT-1 (NCT01297491)II≥2L *Buparlisib30 Sq
33 Nsq
12 wks PFS =
Sq 23.3%, Nsq 20.0%
mOS =
Sq 7.98 mo, Nsq 7.2 mo
ORR =
3.3% Sq, 3.0% Nsq
Closed
at futility
analysis
BASALT-2 (NCT01820325)Ib/II 1LBuparlisib + CBDCA + paclitaxel6 SqORR = 16.7%Early study termination
BASALT-3 (NCT01911325)Ib/II≥2L *Buparlisib
+ docetaxel
27 SqORR 80 mg/daily = 6%
ORR 100 mg/daily = 18%
Early study termination
NCT01487265II≥2L **Buparlisib +
erlotinib
373 mo PFS = 50.4%
mOS = 12.2
ORR = 5.4%
Completed
XL765 (SAR245409, Voxtalisib)NCT01390818IbAny lineVoxtalisib +
pimasertib
(MEK inhibitor)
Advanced solid tumors
(33 NSCLC)
ORR = 5%Completed
NCT00777699I≥2L **Voxtalisib +
erlotinib
Advanced solid tumors
(37 NSCLC)
SD as best responseCompleted
PX-866NCT01204099II2L-3LDocetaxel + PX-866
vs.
docetaxel
95ORR = 6% vs. 0%, p = 0.12
mPFS = 2.0 vs. 2.9 mo,
p = 0.65
mOS = 7.9 vs. 9.4 mo,
p = 0.9
Completed
XL147 (SAR245408)NCT01392924IFurther linesXL147Advanced solid tumors
(24 NSCLC)
ORR = 16.7%Completed
NCT00692640I≥2L **XL147 +
erlotinib
Advanced solid tumors
(20 NSCLC)
ORR = 3.7%
DCR = 51.9%
Completed
GDC-0941
(Pictilisib)
NCT01458067 Ib1LArm A:
pictilisib + CBDCA + paclitaxel
Arm B:
CBDCA + paclitaxel + bevacizumab
Arm C:
CDDP + pemetrexed + bevacizumab
Arm D:
CDDP + pemetrexed
66ORR = 43.9%Completed
Dual PI3K/mTOR inhibitor
GDC-0980 (Apitolisib)NCT01301716 Ib1LArm A:
apitolisib +
CBDCA + paclitaxel
ARM B:
CBDCA + paclitaxel + bevacizumab
Arm C:
CDDP + pemetrexed
Advanced solid
tumors
(39 NSCLC)
ORR = 64%Completed
1L–first line; 2L–second line; 3L–third line; pts–patients; Sq–squamous; Nsq–non-squamous; CBDCA–carboplatin; CDDP–cisplatin; DCR–disease control rate; ORR–objective response rate; mPFS–median progression-free survival; mOS–median overall survival; NSCLC–non-small-cell lung cancer; RP2D–recommended phase 2 dose; EGFR TKI–epidermal growth factor tyrosine kinase inhibitor; MTD–maximum tolerated dose; SD–stable disease; NR–not reached. When patients with solid tumors were included, specific data for NSCLC were reported. * Disease progression to prior platinum-based chemotherapy; ** disease progression to EGFR TKI; *** indicates the line or lines of treatment for advanced NSCLC in which the investigational agent or regimen was employed in each reported trial.
Table 7. Principal single-arm Phase I–II trials on TKI in HER2-positive advanced NSCLC patients.
Table 7. Principal single-arm Phase I–II trials on TKI in HER2-positive advanced NSCLC patients.
DrugTrialPhaseTherapy Line *N ptsMain ResultsStatus
Trastuzumab deruxtecan
(T-DXd)
DESTINY-Lung01
(NCT03505710)
II≥2L42
(HER2exp, HER2mut)
ORR = 61.9%
mPFS = 14 mo
Ongoing
PoziotinibZENITH20-2
(NCT03318939)
II≥2L90
(HER2mut)
ORR = 27.8%
mPFS = 5.5 mo
Ongoing
AfatinibNICHE
(NCT02369484)
II≥2L13
(HER2mut)
ORR = 7.7%
mPFS = 15.9 wks
mOS = 56 wks
Completed
PyrotinibNCT02834936II≥2L60
(HER2mut)
ORR = 31.7%
mPFS = 6.8 mo
mOS = NA
Unknown
Mobocertinib
(TAK-788)
NCT02716116I/II≥1L57
(EGFR exon20ins = 39
HER2 mut = 13)
NAOngoing
Trastuzumab
Emtansine
NCT02675829II≥1L18
(HER2 mut)
ORR = 44%
mPFS = 5 mo
Ongoing
TrastuzumabHOT1303-B
(UMIN000012551)
II≥2L10
(HER2exp, HER2mut)
ORR = 0%
mPFS = 5.2 mo
Completed
DacomitinibNCT00818441II≥2L26ORR = 12%
mPFS = 3 mo
mOS = 9 mo
Completed
1L—first-line; 2L—second-line; N—number, pts—patients; HER2exp—HER2 expressing; HER2mut—HER2 mutated; ORR—overall response rate; wks—weeks; mo—month; mPFS—median progression-free survival; mOS—median overall survival; NA—not available. An asterisk (*) indicates the line or lines of treatment for advanced NSCLC in which the investigational agent or regimen was employed in each reported trial.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Rebuzzi, S.E.; Zullo, L.; Rossi, G.; Grassi, M.; Murianni, V.; Tagliamento, M.; Prelaj, A.; Coco, S.; Longo, L.; Dal Bello, M.G.; et al. Novel Emerging Molecular Targets in Non-Small Cell Lung Cancer. Int. J. Mol. Sci. 2021, 22, 2625. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22052625

AMA Style

Rebuzzi SE, Zullo L, Rossi G, Grassi M, Murianni V, Tagliamento M, Prelaj A, Coco S, Longo L, Dal Bello MG, et al. Novel Emerging Molecular Targets in Non-Small Cell Lung Cancer. International Journal of Molecular Sciences. 2021; 22(5):2625. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22052625

Chicago/Turabian Style

Rebuzzi, Sara Elena, Lodovica Zullo, Giovanni Rossi, Massimiliano Grassi, Veronica Murianni, Marco Tagliamento, Arsela Prelaj, Simona Coco, Luca Longo, Maria Giovanna Dal Bello, and et al. 2021. "Novel Emerging Molecular Targets in Non-Small Cell Lung Cancer" International Journal of Molecular Sciences 22, no. 5: 2625. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22052625

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop