Developments in targeted therapy & immunotherapy—how non-small cell lung cancer management will change in the next decade: a narrative review
Review Article

Developments in targeted therapy & immunotherapy—how non-small cell lung cancer management will change in the next decade: a narrative review

Molly S. C. Li1, Kevin K. S. Mok2, Tony S. K. Mok1

1Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong, China; 2Department of Clinical Oncology, Prince of Wales Hospital, Hong Kong, China

Contributions: (I) Conception and design: All authors; (II) Administrative support: None; (III) Provision of study materials or patients: None; (IV) Collection and assembly of data: MSC Li, KKS Mok; (V) Data analysis and interpretation: MSC Li, KKS Mok; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Molly S. C. Li. Department of Clinical Oncology, The Chinese University of Hong Kong, Prince of Wales Hospital, 30-32 Ngan Shing Street, Shatin, New Territories, Hong Kong, China. Email: molly@clo.cuhk.edu.hk.

Background and Objective: The adoption of targeted therapy and immunotherapy has revolutionised the treatment landscape of non-small cell lung cancer. For early staged disease, incorporation of targeted therapy and immunotherapy has recently been demonstrated to reduce recurrence. Development of targeted therapies in advanced lung cancer is driven by advanced genomic sequencing techniques, better understanding of drug resistance mechanisms, and improved drug designs. The list of targetable molecular alteration is continuously expanding, and next generation molecular therapies have shown promise in circumventing drug resistance. Lung cancer patients may achieve durable disease control with immune checkpoint inhibitors however most patients develop immunotherapy resistance. A wide spectrum of resistance mechanisms, ranging from impaired T-cell activation, presence of coinhibitory immune checkpoints, to immunosuppressive tumour microenvironment, have been proposed. A multitude of novel immunotherapy strategies are under development to target such resistance mechanisms. This review aims to provide a succinct overview in the latest development in targeted therapy and immunotherapy for NSCLC management.

Methods: We searched all original papers and reviews on targeted therapy and immunotherapy in non-small cell lung cancer (NSCLC) using PubMed in June 2022. Search terms included “non-small cell lung cancer”, “targeted therapy”, “immunotherapy”, “EGFR”, “ALK”, “ROS1”, “BRAF V600E”, “MET”, “RET”, “KRAS”, “HER2”, “ERBB2”, “NRG1”, “immune checkpoint”, “PD-1”, “PD-L1”, “CTLA4”, “TIGIT”, “VEGF”, “cancer vaccine”, “cellular therapy”, “tumour microenvironment”, “cytokine”, and “gut microbiota”.

Key Content and Findings: We first discuss the incorporation of targeted therapy and immunotherapy in early staged NSCLC. This includes the latest clinical data that led to the approval of neoadjuvant immunotherapy, adjuvant immunotherapy and adjuvant targeted therapy for early staged NSCLC. The second section focuses on targeted therapy in metastatic NSCLC. The list of targetable alteration now includes but is not limited to EGFR, ALK, ROS1, BRAF V600E, MET exon 14 skipping, RET, KRAS G12C, HER2 and NRG1. Potential drug resistance mechanisms and novel therapeutics under development are also discussed. The third section on immunotherapy in metastatic NSCLC, covers immunotherapy that are currently approved [anti-PD-(L)1 and anti-CTLA4], and agents that are under active research (e.g., anti-TIGIT, cancer vaccine, cellular therapy, cytokine and other TME modulating agents).

Conclusions: This review encompasses the latest updates in targeted therapy and immunotherapy in lung cancer management and discusses the future direction in the field.

Keywords: Non-small cell lung cancer (NSCLC); targeted therapy; immunotherapy


Submitted Sep 07, 2022. Accepted for publication Mar 08, 2023. Published online Mar 23, 2023.

doi: 10.21037/atm-22-4444


Introduction

Background

Lung cancer is the leading cause of cancer death worldwide, claiming 1.8 million lives annually (1). About 85% of lung cancers are non-small cell lung cancer (NSCLC). Patients with lung cancer have dismal prognoses, due to the fact that more than half of them present with metastatic disease, and recurrence is common among those who have early-staged disease (2). Conventional cytotoxic chemotherapy only offers a modest improvement in survival for NSCLC patients (3). The identification of actionable driver oncogenes such as EGFR and ALK, and immune checkpoints such as PD-L1, have led to the development of personalized cancer care. Over the past decade, significant advances in targeted therapy and immunotherapy have transformed the treatment paradigm and survival outcomes of patients with NSCLC. Patients receiving genotype-directed therapy achieve more rapid but durable tumour responses and usually less treatment related toxicities comparing to those who receive conventional chemotherapy. Nonetheless, therapeutic resistance remains a perennial challenge and much research is dedicated to this area.

Rationale and knowledge gap

A myriad of novel therapeutic approaches has shown promise with potential to change future practice. The treatment landscape of NSCLC is rapidly evolving and turning increasingly complex. An updated review in this topic offers oncologists and other clinical practitioners a succinct overview in the state-of-the-art and future trends in NSCLC management.

Objective

This review discusses the latest development in targeted therapy and immunotherapy in NSCLC management. This review also addresses the resistance mechanisms to targeted therapy and immunotherapy and discusses future research direction in this field. We present this article in accordance with the Narrative Review reporting checklist (available at https://atm.amegroups.com/article/view/10.21037/atm-22-4444/rc).


Methods

On June 15th, 2022, a systematic literature search was conducted by MSC Li and KKS Mok. Final approval of literature search was conducted by MSC Li and KKS Mok. An online search of literature utilizing PubMed was employed. Selection criteria included NSCLC, targeted therapy, immunotherapy, EGFR, ALK, ROS1, BRAF, MET, RET, KRAS, HER2, ERBB2, NRG1, immune checkpoint, PD-1, PD-L1, CTLA4, TIGIT, VEGF, cancer vaccine, cellular therapy, tumour microenvironment, cytokine, and gut microbiota from 2005 to 15th June 2022. Only studies in English were included (Table 1).

Table 1

Search strategy summary

Items Specification
Date of search 15th June, 2022
Databases PubMed
Search terms used “non-small cell lung cancer”, “targeted therapy”, “immunotherapy”, “EGFR”, “ALK”, “ROS1”, “BRAF V600E”, “MET”, “RET”, “KRAS”, “HER2”, “ERBB2”, “NRG1”, “immune checkpoint”, “PD-1”, “PD-L1”, “CTLA4”, “TIGIT”, “VEGF”, “cancer vaccine”, “cellular therapy”, “tumour microenvironment”, “cytokine”, and “gut microbiota”
Timeframe 2005 to 15th June, 2022
Inclusion criteria English literature only
Selection process Systematic literature search was conducted by MSC Li and KKS Mok. Final approval of literature search was conducted by MSC Li and KKS Mok

Early staged NSCLC

Historically, neoadjuvant and adjuvant chemotherapy only provide an overall 5% survival benefit (4,5). Motivated by the success of immunotherapy in advanced stage lung cancer (6), much research on immunotherapy has been done in the neoadjuvant or adjuvant setting in recent years to improve this statistic.

Neoadjuvant immunotherapy in surgically resectable NSCLC

The concept of neoadjuvant immunotherapy is supported by the hypothesis that an in-situ tumour may serve as a neoantigen source for stimulation of tumour-specific T-cells (7). Most neoadjuvant clinical trials evaluated pathological response rate as their major study endpoints. While overall survival is the gold standard for study endpoint in neoadjuvant studies, surrogate endpoints are needed for practicality and cost effectiveness. Pathologic response is emerging as one important endpoint because it has strong correlation to recurrence and survival (8), and is more predictive than radiologic response (9).

Earlier trials investigated neoadjuvant immunotherapy with anti-programme death-(ligand)-1 (anti-PD-L1/PD-1) monoclonal antibodies alone. They resulted in major pathologic response rates (MPR, defined as 10% or less viable tumor in resected primary tumour) of 20–45% and pathologic complete response (pCR, defined as no viable tumour cells in resected primary tumour and lymph nodes) rates of 7–10% (7,10-12). A phase II study showed that dual immunotherapy with nivolumab and ipilimumab achieved higher MPR rate (38% versus 22%) and pCR rate (29% versus 9%) compared to nivolumab alone (12).

In general, pathological response rates with single agent immunotherapy were lower than immunochemotherapy combinations. Single arm studies testing neoadjuvant immunochemotherapy combinations reported MPR rates of 57–83% and pCR rates of 18–63%. Toxicity was understandably higher due to the addition of chemotherapy, but surgery was in general feasible (13-15).

The only published phase III trial evaluating chemo-immunotherapy to date was the CheckMate 816 trial, which compared three cycles of neoadjuvant nivolumab plus chemotherapy versus chemotherapy in stage IB (tumor size ≥4 cm) to IIIA patients (AJCC staging 7th edition) without EGFR/ALK alterations. The primary outcomes were met, with an event-free survival (EFS) of 31.6 months versus 20.8 months (HR 0.63, 97.38% CI: 0.43–0.91), pCR of 24% versus 2% (odds ratio 13.94; 99% CI: 3.49–55.75) in the chemoimmunotherapy versus chemotherapy arms respectively. Patients with high PD-L1 expression (HR 0.24, 95% CI: 0.10–0.61) or stage III disease (HR 0.54, 95% CI: 0.37–0.80) benefited more from the neoadjuvant immunochemotherapy combination. The trial also validated that pCR was strongly predictive of EFS (16). A number of phase III neoadjuvant immunochemotherapy studies are underway (Table 2).

Table 2

Key randomized perioperative immunotherapy studies in early staged NSCLC

Study name Intervention Primary endpoint Accrual status NCT identifier
Neoadjuvant
   CheckMate 816 Nivolumab plus platinum based chemotherapy pCR, EFS Results published NCT02998528
   CheckMate 77T Nivolumab plus platinum based chemotherapy followed by adjuvant nivolumab EFS Recruiting NCT04025879
   KEYNOTE-671 Pembrolizumab plus platinum based chemotherapy followed by adjuvant pembrolizumab EFS, OS Active, not recruiting NCT03425643
   IMpower030 Atezolizumab plus platinum based chemotherapy followed by adjuvant atezolizumab EFS Active, not recruiting NCT03456063
   AEGEAN Durvalumab plus platinum based chemotherapy pCR, EFS Recruiting NCT03800134
Tislelizumab plus platinum based chemotherapy followed by adjuvant tislelizumab MPR, EFS Recruiting NCT04379635
Sintilimab plus platinum based chemotherapy followed by adjuvant sintilimab pCR, EFS Not yet recruiting NCT05116462
SHR-1316 plus platinum based chemotherapy followed by adjuvant SHR-1316 MPR, EFS Recruiting NCT04316364
Toripalimab plus platinum based chemotherapy followed by adjuvant toripalimab MPR, EFS Recruiting NCT04158440
Adjuvant
   IMpower010 Atezolizumab (after adjuvant chemotherapy) DFS Results published NCT02486718
   KEYNOTE-091 Pembrolizumab (after adjuvant chemotherapy but chemotherapy not mandatory) DFS Results published NCT02504372
   ANVIL Nivolumab (after adjuvant chemotherapy) DFS, OS Active, not recruiting NCT02595944
   BR31 Durvalumab (after adjuvant chemotherapy but chemotherapy not mandatory DFS in PD-L1 TC ≥25% Active, not recruiting NCT02273375
   LungMate-008 Toripalimab plus platinum based chemotherapy for 4 cycles DFS Not yet recruiting NCT04772287
   NADIM-ADJUVANT Nivolumab plus carboplatin plus paclitaxel followed by maintenance nivolumab DFS Recruiting NCT04564157
   ALCHEMIST-IO Arm 1: adjuvant chemotherapy alone followed by maintenance pembrolizumab
Arm 2: pembrolizumab plus platinum based chemotherapy followed by maintenance pembrolizumab
DFS Recruiting NCT04267848
   MERMAID-1 Durvalumab plus platinum based chemotherapy DFS in MRD+ analysis set Active, not recruiting NCT04385368
   MERMAID-2 Durvalumab DFS in PD-L1 TC ≥1% Active, not recruiting NCT04642469

NSCLC, non-small cell lung cancer; DFS, disease-free survival; EFS, event-free survival; MPR, major pathological response; MRD, minimal residual disease; OS, overall survival; pCR, pathological complete response; PD-L1, programme death-ligand 1; TC, tumour cells.

Future studies will explore combining neoadjuvant immunotherapy with antiangiogenic drugs such as lenvatinib (NCT04875585), apatinib (NCT04379739) and bevacizumab (NCT04973293), or relatlimab (NCT04205552), an anti-LAG3 checkpoint inhibitor. Tri-modality combinations with chemotherapy, immunotherapy, and radiotherapy are also being studied (NCT04245514, NCT05157542).

Adjuvant immunotherapy in surgically resected NSCLC

IMpower010 was the first published phase III immunotherapy study in the adjuvant setting in surgically treated lung cancer, comparing adjuvant atezolizumab up to one year versus supportive care after surgery and adjuvant chemotherapy (Table 2). This study showed improved disease-free survival (DFS) in stage II to IIIA patients with PD-L1 ≥1% (HR 0.66, 95% CI: 0.50–0.88; 3-year DFS 60% versus 48%) and in all comers of stage II to IIIA (HR 0.79, 95% CI: 0.64–0.96; 3-year DFS 56% versus 49%). Exploratory analysis suggested that the PD-L1 expression might be related to outcome, with a HR of 0.43 (95% CI: 0.27–0.68) in the ≥50%, but 0.97 (95% CI: 0.72–1.31) in the ≤1% group (17).

Data from the second interim analysis for adjuvant pembrolizumab was presented recently. The KEYNOTE-091 study was a triple blinded phase III randomized study comparing adjuvant pembrolizumab versus placebo for stage IB (≥4 cm) to IIIA patients (AJCC staging 7th edition) after surgery with or without chemotherapy. The study reported a significant DFS benefit with pembrolizumab but unlike IMpower010, the DFS outcomes did not correlate with PD-L1 expression. The reason for this discrepancy in outcome was not clear (18). Other phase III trials testing adjuvant PD-(L)1 blockade in patients with resected NSCLC have completed accrual and will provide additional insight into the potential biomarkers (Table 2). More importantly, in future, testing clinically whether there are differences in outcome between a neoadjuvant and adjuvant approach should be further pursued.

Immunotherapy in locally advanced unresectable lung cancer

Definitive chemoradiotherapy followed by consolidation durvalumab for up to 1 year is now the standard for unresectable stage III NSCLC based on the landmark PACIFIC trial (19). In the latest update, durvalumab versus placebo was associated with median overall survival (OS) of 47.5 versus 29.1 months. Patients with PD-L1 expression ≤1% or EGFR/ALK alteration did not yield OS benefit from the study drug [HR 1.15 (95% CI: 0.75–1.75) and HR 0.85 (95% CI: 0.37–1.97) respectively] (20).

Building upon the PACIFIC trial, the COAST trial was a phase II trial incorporating oleclumab (anti-CD73 mAB) or monalizumab (anti-NKG2A mAB), novel drugs with immunomodulatory effects, to standard consolidation durvalumab. Encouraging response rates of 30.0% (oleclumab plus durvalumab), 35.5% (monalizumab plus durvalumab), versus 17.9% (durvalumab) were reported (21). This has now moved onto a phase III study (NCT05221840).

Integrating immunotherapy into chemoradiotherapy was feasible in a phase II trial involving pembrolizumab with response rate of about 70% and Grade 3 pneumonitis of 7–8% (22). Another similar trial involving concurrent durvalumab is underway (NCT03519971). Novel immunotherapy agents such as anti-TIGIT are being investigated as a component of consolidation therapy (NCT04513925, NCT05211895) and as part of the chemoradiotherapy regime (NCT04866017).

Neoadjuvant and adjuvant therapy in NSCLC with EGFR or other driver oncogene

Neoadjuvant tyrosine kinase inhibitor (TKI) is not yet an established standard treatment for patients with NSCLC harboring driver mutation. The EMERGING-CTONG 1103 trial compared neoadjuvant erlotinib with neoadjuvant chemotherapy in stage IIIA N2 patients with EGFR exon 19 or 21 mutations. While objective response (54.1% versus 34.3%, P=0.09) was numerically higher and progression free survival (PFS) was statistically significantly better of 21.5 months versus 11.4 months (P<0.001), neoadjuvant erlotinib did not improve overall survival (median OS 42.2 versus 36.9 months, HR 0.83, P=0.513) (23). This trial raised an important message that PFS benefit in an neoadjuvant targeted therapy study does not necessarily translate into OS benefit. NEOS, a single arm trial of 18 patients, reported response rate of 73.3% with neoadjuvant osimertinib (24). An ongoing phase III trial called NEOADAURA will determine whether osimertinib with or without chemotherapy is beneficial in the neoadjuvant setting (NCT04351555).

The most promising data on adjuvant epidermal growth factor receptor (EGFR) TKI involved osimertinib in the ADAURA trial. This was a phase III trial in which patients with resected stage IB to IIIA NSCLC (AJCC staging 7th edition) with EGFR exon 19 deletion or L858R mutation were randomized to receive either up to three years of osimertinib or placebo. The reported 24-month DFS was an impressive 90% versus 44% (HR 0.17, P<0.001). Given the overwhelming DFS gain the study was unblinded early and the drug was Food and Drug Administration (FDA) approved for this indication. OS data has yet to mature (25).

Clinical data on perioperative TKI for NSCLC harboring other driver oncogenes (e.g., ALK, ROS1) is scarce due to rarity of these tumours. ALINA is an ongoing phase III study comparing adjuvant alectinib with standard platinum-based chemotherapy in patients with resected ALK positive NSCLC (NCT0456076). NAUTIKA1 is a phase II study in which patients receive genotype-directed, neoadjuvant TKI followed by surgery, adjuvant chemotherapy and 2 years of TKI (NCT04302025). Primary study outcome is MPR rate. At present, osimertinib is the only FDA approved targeted therapy for early staged NSCLC harboring a driver mutation.

Early-staged NSCLC: future direction

The phase III studies presented above have set the stage for immunotherapy and targeted therapy in early staged NSCLC. Future OS data and results of the other ongoing phase III studies will confirm the benefit of immunotherapy and targeted therapy in this setting. The classical notion that the histological subtype and molecular profile are irrelevant in early staged NSCLC management has become obsolete. EGFR mutation and PD-L1 expression should be tested at diagnosis to guide treatment plan. Multidisciplinary discussion is crucial for management of stage III, or even stage II NSCLC as different treatment approaches are now viable. Given the improving response rates of neoadjuvant treatment, tumours that are determined unresectable at diagnosis may be converted to resectable. As a result, the line between unresectable and resectable tumours may blur. Future studies will also focus on identifying patients, for example those who fail to achieve pCR, or those with minimal residual disease after surgery, who will benefit from intensive treatment.


Targeted therapy in metastatic NSCLC

Overview

The foundation of targeted therapy in lung cancer is based on the knowledge that certain genetic alterations act as primary drivers of cancer formation, such that inhibition of the driver oncogene leads to cancer growth arrest and cell death. Targeted therapies in NSCLC are conventionally referred to small molecule gene-specific tyrosine kinase inhibitors. Recently, innovations in drug designs have led to development of newer classes of targeted therapies like antibody-drug conjugates (ADC) and bispecific antibodies. Tyrosine kinase inhibitors (TKIs) have proven enormous success in NSCLC treatment as up to one half of lung adenocarcinomas (LUAD) harbour a driver oncogene (26-28) (Figure 1). To date, there are nine oncogenes in NSCLC with FDA approved therapies.

Figure 1 Prevalence of actionable driver alterations in lung adenocarcinoma. Frequency of driver oncogenes in LUADs in Caucasians, adapted from Jordan et al. (28). LUADs, lung adenocarcinomas.

EGFR mutation

Currently, 5 EGFR TKIs (namely erlotinib, gefitinib, afatinib, dacomitinib, and osimertinib) are approved for treatment naïve, advanced EGFR mutated NSCLC (Table 3) (29-41). The FLAURA study randomized 556 patients with untreated advanced EGFR-mutated NSCLC to receive osimertinib or a first-generation EGFR TKI. Osimertinib demonstrated superiority over first-generation TKI in terms of PFS, OS, intracranial objective response rate (ORR), as well as a more favourable side effect profile, and thus is the preferred first-line treatment option for advanced NSCLC harbouring a sensitizing EGFR mutation (42,43).

Table 3

Key randomized targeted therapy studies in NSCLC with actionable genomic alterations

Study name Intervention Comparator Treatment line Median PFS, months, HR (95% CI) Median OS, months, HR (95% CI) Ref
EGFR
   IPASS Gefitinib Carboplatin plus paclitaxel 1st 9.5 vs. 6.3, HR 0.48 (0.36 to 0.64) 18.8 vs. 17.4, HR 1.0 (0.76 to 1.33) (29,30)
   NEJ002 Gefitinib Carboplatin plus paclitaxel 1st 10.8 vs. 5.4, HR 0.30 (0.22 to 0.41) 30.5 vs. 23.6, HR 0.887 (0.63–1.24) (31,32)
   EURTAC Erlotinib Platinum based chemotherapy 1st 9.7 vs. 5.2, HR 0.37 (0.25–0.54) 19.3 vs. 19.5, HR 1.04 (0.65–1.68) (33)
   OPTIMAL Erlotinib Carboplatin plus gemcitabine 1st 13.1 vs. 4.6, HR 0.16 (0.10–0.26) 22.8 vs. 27.2, HR 1.19 (0.83–1.71) (34,35)
   LUX-LUNG-7 Afatinib Gefitinib 1st 11.0 vs. 10.9, HR 0.73 (0.57–0.95) 27.9 vs. 24.5, HR 0.86 (0.66–1.12) (36,37)
   ARCHER-1050 Dacomitinib Gefitinib 1st 14.7 vs. 9.2, HR 0.59 (0.47–0.74) 34.1 vs. 26.8, HR 0.76 (0.58 to 0.99) (38,39)
   AURA3 Osimertinib Platinum plus pemetrexed 2nd, progression after first-line EGFR TKI with T790M mutation 10.1 vs. 4.4, HR 0.30 (0.23 to 0.41) 26.8 vs. 22.5, HR 0.87 (0.67 to 1.12) (40,41)
   FLAURA Osimertinib Erlotinib or gefitinib 1st 18.9 vs. 10.2, HR 0.46 (0.37 to 0.57) 38.6 vs. 31.8, HR 0.80 (0.64 to 1.00) (42,43)
ALK
   PROFILE-1014 Crizotinib Platinum plus pemetrexed 1st 10.9 vs. 7.0, HR 0.45 (0.35 to 0.60) NR vs. 47.5, HR 0.76 (0.548 to 1.053) (44,45)
   ASCEND-4 Ceritinib Platinum plus pemetrexed 1st 16.6 vs. 8.1, HR 0.55 (0.42 to 0.73) NR vs. 26.2, HR 0.73 (0.50 to 1.08) (46)
   ALEX Alectinib Crizotinib 1st 34.8 vs. 10.9, HR 0.43 (0.32 to 0.58) NR vs. 57.4, HR 0.67 (0.46 to 0.98) (47)
   ALTA-1L Brigatinib Crizotinib 1st 24.0 vs. 11.1, HR 0.48 (0.35 to 0.66) NR vs. NR, HR 0.81 (0.51 to 1.22) (48)
   eXalt3 Ensartinib Crizotinib 1st 25.8 vs. 12.7, HR 0.51 (0.35 to 0.72) NR vs. NR, HR 0.91 (0.54 to 1.54) (49)
   CROWN Lorlatinib Crizotinib 1st NR vs. 9.3, HR 0.28 (0.19 to 0.41) NR vs. NR, HR 0.72 (0.41 to 1.25) (50)

NSCLC, non-small cell lung cancer; ALK, anaplastic lymphoma kinase; EGFR, epidermal growth factor receptor; HR, hazard ratio; NR, not reached; OS, overall survival; PFS, progression free survival; Ref, reference.

Treatment strategy upon osimertinib resistance is under active research. The standard treatment for patients after osimertinib resistance is cytotoxic chemotherapy. With growing understanding of the resistance patterns to osimertinib and advances in drug designs, multiple new treatment approaches have shown promise.

EGFR exon 20 C797S mutation is the most common EGFR-dependent mechanism of osimertinib resistance (7–26%) and interferes with drug binding to the EGFR protein (51). Preclinical data showed that fourth generation EGFR TKIs, such as BLU-701, BLU-945, BBT-176 and JBJ-09-063, were active against C797S resistance (52-55). Phase I/II studies are now underway to study the clinical efficacy and safety of these agents (e.g., NCT 05153408, NCT04820023).

MET amplification accounts for up to 20% of osimertinib resistance (51). The combination of EGFR TKI plus a selective MET inhibitor (e.g., tepotinib, capmatinib, savolitinib) was tested in patients with MET overexpression or amplification resistance in several phase Ib/II trials. Reported ORRs were around 30–60% supporting TKI combination as a treatment option for patients with acquired MET dysregulation (56-59). Phase III randomized studies such as GEOMETRY-E (NCT04816214) and SAFFRON (NCT-05261399) will compare such combinations with standard platinum-based chemotherapy. EGFR TKI resistance mediated by other actionable alterations are rare but treatment success of different TKI combinations have been reported in literature (60-62), highlighting the importance of repeating genomic profiling after TKI failure to search for druggable resistance mutations.

Amivantamab is a bispecific antibody against EGFR and c-MET (63). In the CHRYSALIS and CHRYSALIS-2 single arm phase II studies, the combination of amivantamab and lazertinib (a third-generation EGFR TKI) achieved response rates of 36% and 32% respectively in patients who progressed on osimertinib (64,65). These data supported bispecific antibody to be an alternative approach other than tyrosine kinase inhibition in targeting oncogene-addicted NSCLCs by induction of antibody-dependent cellular cytotoxicity (ADCC) and simultaneous inhibition of multiple signalling pathways (66).

HER3 is typically overexpressed on EGFR-mutated NSCLC (67) and patritumab deruxtecan (HER3-DXd) is an ADC against HER3. In a phase I study, ORR was 39% and median PFS was 8.2 months among 57 patients who had prior EGFR TKI and platinum-based chemotherapy and received HER3-DXd (68). The HERTHENA-Lung02 is a phase III study (NCT05338970) comparing HER3-DXd with platinum-based chemotherapy for patients with EGFR-mutated NSCLC failing osimertinib.

ALK rearrangement

Second and third generation ALK TKIs (namely ceritinib, alectinib, brigatinib, ensartinib, lorlatinib) have largely supplanted crizotinib for treatment-naïve, advanced ALK-rearranged NSCLC as all of them demonstrated superior PFS comparing with crizotinib in the upfront setting, except that ceritinib was compared with chemotherapy. (Table 3) (44-46,48-50,69). Although cross-trial comparison suggested that lorlatinib offered more durable disease control compared to the second generation ALK TKIs (3-year PFS 63% with lorlatinib, 45% with alectinib and 43% with brigatinib) (47,48,70), the optimal first-line treatment is still an open debate as lorlatinib usually remains effective after second generation ALK TKI failure (71).

Resistant ALK mutations most often occur in the gatekeeper region (e.g., L1196M and G1269A) or the solvent front region (e.g., G1202R) after first and second generation ALK TKI treatment (72). Serial ALK TKI treatment potentially predisposes to multiple resistant kinase mutations and lorlatinib resistance (73). Fourth generation ALK TKIs like TPX-0131 are designed to bind precisely to the adenine binding site of ATP to avoid clashing with the solvent front, hinge or gatekeeper area, and thus may overcome double- or triple-mutant resistant kinase mutations (74). TPX-0131 and NVL-655 are now being evaluated in phase I clinical studies which involve patients who have failed multiple lines of ALK TKI including lorlatinib (NCT04849273, NCT05384626).

ROS1 rearrangement

Crizotinib and entrectinib are approved therapies for ROS1-rearranged NSCLC. PROFILE 1001 was a single arm phase II study in which 50 patients with ROS1-rearranged NSCLC received crizotinib. Crizotinib achieved an ORR of 72% and a median PFS of 19.2 months (75). In the integrated analysis of three entrectinib studies (ALKA-372-001, STARTRK-1, and STARTRK-2), entrectinib achieved a similar ORR of 77% and a median PFS of 19.0 months (76). Entrectinib may be preferred in patients with brain metastases in view of its excellent intracranial activity (intracranial ORR 52.2%) (77). Besides crizotinib and entrectinib, ceritinib and lorlatinib are also both active in ROS1-rearranged NSCLC but are not FDA approved (78,79). ROS1G2032R solvent-front mutation is the most common resistance mechanism following crizotinib progression and confers resistance to all commercially available ROS1 inhibitors (80). Interim data of the TRIDENT-1 phase I/II study showed that repotrectinib (TPX-005), a next generation ROS1/TRK inhibitor, was active against ROS1 TKI-naïve and pretreated patients (81). Enrollment of this study is now ongoing (NCT03093116).

MET exon 14 skipping

Three highly selective type IB MET inhibitors, namely capmatinib, tepotinib, and savolitinib, were proven highly efficacious in patients with advanced MET exon 14 skipping positive (METex14) NSCLC. In GEOMETRY mono-1, treatment naïve patients with advanced METex14 NSCLC achieved an ORR of 68% and median PFS of 12.4 months with capmatinib. In the pretreated cohort, ORR was 41% and median PFS was 5.4 months (82,83). In the VISION study, 152 patients with NSCLC harbouring METex14, either detected by tissue or liquid biopsy, received tepotinib. Response rates were similar at 50% either in the tissue-biopsy or liquid-biopsy group, regardless of number of lines of previous treatment (84). Both drugs exhibited intracranial antitumour efficacy (84,85). Savolitinib was evaluated in 70 patients with pretreated METex14 NSCLC in China. Notably, 25 patients (36%) had pulmonary sarcomatoid carcinoma (PSC), which was underrepresented in the GEOMETRY mono-1 and the VISION study (<8% of patients had PSC). ORR in the intention-to-treat population and PSC subgroup were 49% and 40% respectively (86). Peripheral oedema is the major toxicity from MET inhibitors. Currently, capmatinib and tepotinib are FDA approved and savolitinib is approved in China for advanced METex14 NSCLC. Multiple treatment strategies targeting MET-dysregulated NSCLC, such as type II MET inhibitor (e.g., merestinib, NCT02920996), MET-based ADC (e.g., telisotuzumab vedotin, NCT03539536) and bispecific antibody (e.g., amivantamab, NCT02609776), are under study.

RET rearrangement

Recently, two selective RET inhibitors, demonstrated high efficacy in RET rearranged NSCLC and received FDA approvals for this treatment indication. Selpercatinib was tested in 144 patients with RET-rearranged NSCLC in the LIBRETTO-001 multicentre single arm phase II study. In the treatment-naïve cohort, ORR was 85% and median PFS was not reached at data cutoff. In the platinum pretreated cohort, ORR was 64% and median PFS was 16.5 months (87). The ARROW study evaluated 121 patients with RET-rearranged NSCLC who received pralsetinib. ORR was 70% in the treatment naïve cohort and 61% in the pretreated cohort (88). Grade 3 and 4 toxicities due to RET inhibitors were rare and mainly included hypertension, deranged liver function, and cytopenia. The LIBRETTO-431 (NCT04194944) and ACCELE-RET (NCT04222972) are comparing selpercatinib and pralsetinib respectively, with platinum-based chemotherapy with or without pembrolizumab in patients with treatment-naïve advanced RET-rearranged NSCLC. Novel RET inhibitors (e.g., TPX-0046, NCT04161391; BOS172738, NCT03780517) designed to overcome acquired RET resistant mutations are now evaluated in phase I/II clinical trials (89,90).

BRAF V600E mutation

The combination of dabrafenib and trametinib is FDA approved for the treatment of advanced, BRAF V600E mutated NSCLC. Clinical efficacy achieved by the BRAF and MEK inhibitor combination (ORR 64% for both untreated and treatment naïve patients, median PFS 14.6 months for untreated cohort and 8.6 months for pretreated cohort) was considerably higher than BRAF inhibitor monotherapy (ORR about 40% and median PFS about 5–6 months) (91-93). Pyrexia is a specific and common (up to 50%) side effect from the drug combination which is usually ameliorated by antipyretics and dose adjustment.

NTRK rearrangement

Two TRK inhibitors, larotrectinib and entrectinib, received FDA approval as tumor agnostic therapies for patients with tumours harbouring NTRK fusions. In an updated integrated analysis of three phase I/II clinical trials which evaluated larotrectinib in 159 patients with NTRK fusion-positive solid tumours, ORR was 79% and median PFS was 25.8 months (94). Another integrated analysis of three phase I/II clinical trials analysed the clinical efficacy of entrectinib in 121 patients with NTRK fusion-positive tumours. The ORR was 61% and the median PFS was 13.8 months (95). Both larotrectinib and entrectinib showed high efficacy across a wide range of tumour types including NSCLC. TRK inhibitors are associated with specific on-target side events like cognitive impairment, dizziness, weight gain and drug withdrawal pain but drug discontinuation due to treatment-related adverse events is rare (<10%). In a small-scale study of 18 patients who progressed on first generation TRK inhibitors (larotrectinib or entrectinib), a resistant solvent front mutation was identified in 13 of them (96). Preclinical data showed that next generation TRK inhibitors such as taletrectinib, repotrectinib, and selirectinib, were effective against NTRK fusion positive tumours with secondary resistant NTRK mutations (81,97,98).

KRAS G12C mutation

KRAS was considered undruggable in the past due to the absence of an identifiable drug binding pocket on KRAS protein and its strong affinity to ATP rendering design of KRAS inhibitors inherently difficult (99). In 2013, the Shokat lab identified a small pocket adjacent to the switch-II region that was only present in GDP-bound, G12C mutant KRAS protein. The research team designed a small, KRAS G12C allele-specific molecule (ARS-1620) that could bind to this pocket and lock the KRAS protein in its inactive, GDP-bound state (100). Non-cancer cells that do not carry the KRAS G12C mutation could be spared from the toxicities, resulting in a much wider therapeutic index. Further refinement in drug structure designs have led to development of molecules with enhanced potency that are tested in clinic today.

Sotorasib (AMG510) was the first KRAS G12C inhibitor adopted in clinic setting. In CodeBreak 100 study, the drug was evaluated in 126 patients with pretreated KRAS G12C mutated NSCLC. The ORR was 37% and the median PFS was 6.8 months (101). Recently, the results of the phase III CodeBreak 200 trial were published. Sotorasib achieved superior PFS and ORR compared to docetaxel in patients who progressed after prior platinum-based chemotherapy and immune checkpoint inhibitor (102). Another KRAS G12C inhibitor, adagrasib (MRTX849), achieved treatment response rate of 43% and median PFS 6.5 months in the KRYSTAL-1 phase I-II study enrolling 116 patients (103). A number of KRAS G12C inhibitors, such as JDQ443, JAB-21822, and GDC-6036, are under clinical investigation (NCT05132075, NCT05276726, NCT04449874).

A heterogenous spectrum of resistance mechanisms have been identified in the setting of KRAS G12C inhibitor resistance, of which secondary KRAS mutations or alterations in RTK-RAS signalling pathway are most prevalent (104,105). These data supported the approach of combining a KRAS G12C inhibitor with an RTK or SHP2 inhibitor in order to achieved more sustained RAS inhibition (NCT04330664, NCT04185883).

Targeting KRAS mutants other than G12C is more challenging given the lack of a drug binding pocket and even higher affinity to GTP. Nonetheless, preclinical data on KRAS G12D, KRAS G12V, and pan-KRAS inhibitors is emerging (106-110).

EGFR exon 20 insertion

EGFR exon 20 insertion (EGFRex20ins) is well known to be EGFR TKI resistant due to its unique steric configuration. Recently, two drugs were approved by the FDA for treatment of advanced EGFRex20ins NSCLC in the second line setting. Mobocertinib is an oral TKI designed specifically targeting EGFRex20ins. The drug achieved an ORR of 28% and median PFS of 7.3 months in platinum-pretreated patients with advanced EGFRex20ins NSCLC (111). Amivantamab is a bispecific EGFR-cMET antibody achieving a tumour response rate of 40% and median PFS of 8.3 months in 81 patients in the CHRYSALIS study (112).

HER2 mutation

Studies in the past targeting HER2 mutation have yielded disappointing results. Various classes of anti-HER2 agents, including monoclonal antibodies, TKIs, and ADCs, have been investigated in HER2-mutated NSCLC. Recently, a few agents have demonstrated signals of antitumour activity (113-116). Among all anti-HER2 targeted therapies, trastuzumab deruxtecan (T-DXd) has shown the best promise. Compared to trastuzumab emtansine (T-DM1), T-DXd has a different cytotoxic payload and an enhanced linker-payload system enabling a higher drug-to-antibody ratio (1 to 8 for T-DXd compared to 1 to 4 for T-DM1) while maintaining high stability in plasma (117). Recently, the results of DESTINY-Lung01 trial were published. Total of 91 patients with HER2-mutated NSCLC were enrolled into the study. T-DXd achieved an ORR of 55%, DCR of 92% and median PFS of 8.2 months. Interstitial lung disease is a specific adverse event of T-DXd that occurred in a quarter of patients in this study (118). In August 2022, the FDA granted accelerated approval to T-DXd for patients with advanced HER2-mutated NSCLC failing first-line therapy. The upcoming DESTINY-Lung04 study will evaluate T-DXd versus standard of care in patients with untreated, advanced NSCLC harbouring a HER2 mutation (NCT05048797).

NRG1 rearrangement

NRG1-rearrangement represents a rare but targetable oncogene in NSCLC (0.2%) and causes cancer by inducing ErbB2-ErbB3 dimerization. Zenocutuzumab, a bispecific HER2/HER3 antibody, and seribantumab, an anti-HER3 monoclonal antibody, showed encouraging signal in targeting NRG1-rearranged solid tumours including NSCLC (119-121). Both drugs are now being studied in phase II studies (NCT02912949, NCT04383210).

Antibody-drug-conjugates under development

TROP2 is ubiquitously expressed on lung cancer cells and thus a favourable target for development of ADC (120). Sacituzumab govitecan is a TROP-2 ADC connected to an irinotecan derivative payload SN-38. In a single arm cohort enrolling 54 heavily pretreated patients, ORR was 17% (122). Datopotamab deruxtecan (Dato-DXd) is another TROP-2 ADC using deruxtecan as the cytotoxic payload. In the NSCLC cohort of the TROPION-PanTumor01 Phase I study, ORR was 24%. Interstitial lung disease was present in 11% of patients (123). Dato-DXd was also active in patients with actionable genetic alterations (124). Tusamitamab ravtansine, an anti-CEACAM5 ADC, has shown antitumour efficacy in NSCLC. Tumour response rate was 20% in a heavily pretreated population with CEACAM5 highly expressed NSCLC (125). These drugs are now compared with docetaxel in the platinum-refractory setting (NCT04656652, NCT05089734, NCT04154956), and combinations of TROP-2 ADC with immunotherapy are also explored (NCT04526691, NCT04612751, NCT05186974).

Targeted therapy: future direction

Advances in structural bioinformatics and computational biology have led to drug discoveries that target “undruggable” alterations such as KRAS and EGFRex20ins. Consequently, targeted therapies are no longer limited to TKIs and monoclonal antibodies, but also comprise allosteric inhibitors, ADCs and bispecific antibodies. Increasing adoption of high throughput genomic sequencing as well as liquid biopsy testing has facilitated detection of rare genetic alterations and depiction of drug resistance mechanisms. Ongoing research focuses on characterization of TKI resistance mechanisms and development of novel therapeutic approaches targeting actionable resistance alterations. As a result, molecular profiling is not only essential at the time of diagnosis, but it may be clinically relevant to track the evolution and genomic changes of the tumour during the treatment course in order to establish a personalized therapeutic strategy.


Immunotherapy in metastatic NSCLC

PD-L1 blockade

Immune checkpoint inhibitor (ICI), notably anti-PD-L1/PD-1 antibody, is the cornerstone of immunotherapy for multiple cancer types including NSCLC. PD-1/PD-L1 binding leads to intratumoral T-cell exhaustion thus blocking this interaction may reactivate exhausted T-cells for cancer cell killing. PD-(L)1 blockade is now considered standard of care for patients with advanced NSCLC without actionable oncogenic alterations. At present, multiple anti-PD-(L)1 agents have been approved for metastatic NSCLC, either as monotherapy or in combination with other drugs (Figure 2). Treatment efficacy of anti-PD-(L)1 directly correlates with PD-L1 expression on tumour cells. For patients with treatment naïve, EGFR/ALK negative NSCLC with PD-L1 tumour proportion score (TPS) ≥50%, first-line anti-PD-(L)1 monotherapy was superior to chemotherapy in terms of PFS and OS (126-128) (Table 4) In the KEYNOTE-024 study, five-year overall survival was 31% for patients assigned to the pembrolizumab arm (129). Pembrolizumab monotherapy is approved but generally not recommended for patients with NSCLC with PD-L1 TPS 1–49%, as subgroup analysis showed no survival benefit over chemotherapy (127,130).

Figure 2 Timeline of FDA approval of immunotherapy in metastatic NSCLC. PD-L1, programme death-ligand 1; NSCLC, non-small cell lung cancer; non-SqCC, non-squamous.

Table 4

Key randomized immunotherapy trials in treatment naïve, EGFR/ALK-ve, NSCLC

Study name Intervention Comparator Key inclusion criteria Median PFS, months, HR (95% CI) Median OS, months, HR
(95% CI)
Ref
Anti-PD-(L)1 single agent
   KEYNOTE-024 Pembrolizumab Platinum based chemotherapy NSCLC PD-L1 TPS ≥50% 10.3 vs. 6.0, HR 0.50 (0.37 to 0.68) 30.0 vs. 14.2, HR 0.63 (0.47 to 0.86) (126,129)
   Impower110 Atezolizumab Platinum based chemotherapy NSCLC PD-L1 ≥50% of tumour cells or ≥10% of tumour-infiltrating immune cells 8.1 vs. 5.0, HR 0.63 (0.45 to 0.88) 20.2 vs. 13.1, HR 0.59 (0.40 to 0.89) (127)
   EMPOWER-Lung 1 Cemiplimab Platinum based chemotherapy NSCLC PD-L1 TPS ≥50% 8.2 vs. 5.7, HR 0.54 (0.43 to 0.68) NR vs. 14.2, HR 0.57 (0.42 to 0.77) (128)
   KEYNOTE-042 Pembrolizumab Platinum based chemotherapy NSCLC PD-L1 TPS ≥1% 5.4 vs. 6.5, HR 1.07 (0.94 to 1.21) 16.7 vs. 12.1, HR 0.81 (0.71 to 0.93) (130)
Anti-PD-(L)1 plus chemotherapy combination
   KEYNOTE-189 Pembrolizumab-platinum-pemetrexed Platinum plus pemetrexed Non squamous, any PD-L1 TPS 9.0 vs. 4.9, HR 0.48 (0.40 to 0.58) 22.0 vs. 10.7, HR 0.56 (0.45 to 0.70) (131,132)
   KEYNOTE-407 Pembrolizumab-platinum based chemotherapy Platinum based chemotherapy Squamous, any PD-L1 TPS 6.4 vs. 4.8, HR 0.56 (0.56 to 0.70) 15.9 vs. 11.3, HR 0.64 (0.49 to 0.85) (133)
   IMpower150 Atezolizumab-bevacizumab-carboplatin-paclitaxel Bevacizumab-carboplatin-paclitaxel Non-squamous, any PD-L1 TPS 8.3 vs. 6.8, HR 0.62 (0.52 to 0.75) 19.2 vs. 14.7, HR 0.78 (0.64 to 0.96) (134)
   IMpower132 Atezolizumab-platinum-pemetrexed Platinum plus pemetrexed Non squamous, any PD-L1 TPS 7.6 vs. 5.2, HR 0.60 (0.49 to 0.72) 17.5 vs. 13.6, HR 0.86 (0.71 to 1.06) (135)
   IMpower130 Atezolizumab-carboplatin- nab-paclitaxel Carboplatin plus nab-paclitaxel Non squamous, any PD-L1 TPS 7.0 vs. 5.5, HR 0.64 (0.54 to 0.77) 18.6 vs. 13.9, HR 0.79 (0.64 to 0.98) (136)
   IMpower131 Atezolizumab-carboplatin- nab-paclitaxel Carboplatin plus nab-paclitaxel Squamous, any PD-L1 TPS 6.3 vs. 5.6, HR 0.71 (0.60 to 0.85) 14.2 vs. 13.5, HR 0.88 (0.73 to 1.05) (137)
   ORIENT-11 Sintilimab-platinum-pemetrexed Platinum plus pemetrexed Non squamous, any PD-L1 TPS 8.9 vs. 5.0, HR 0.482 (0.362 to 0.643) NR vs. NR, HR 0.609 (0.400 to 0.926) (138)
   ORIENT-12 Sintilimab-platinum-gemcitabine Platinum plus gemcitabine Squamous, any PD-L1 TPS 5.1 vs. 4.9, HR 0.621 (0.473 to 0.815) Not reached (139)
   RATIONALE 304 Tislezumab-platinum-pemetrexed Platinum plus pemetrexed Non squamous, any PD-L1 TPS 9.7 vs. 7.6, HR 0.645 (0.462 to 0.902) Not reached (140)
   RATIONALE 307 Tislezumab-carboplatin-paclitaxel (Arm A) Carboplatin-paclitaxel (Arm C) Squamous,
any PD-L1 TPS
7.6 vs. 5.5, HR 0.524 (0.370 to 0.742) Not reached (141)
Tislezumab-carboplatin- nab-paclitaxel (Arm B) 7.6 vs. 5.5, HR 0.478 (0.336 to 0.679) Not reached
   CameL Camrelizumab-carboplatin-pemetrexed Carboplatin-pemetrexed Non-squamous, any PD-L1 TPS 11.3 vs. 8.3, HR 0.60 (0.45 to 0/79) NR vs. 20.9, HR 0.73 (0.53 to 1.02) (142)
   CameL-Sq Camrelizumab-carboplatin-paclitaxel Carboplatin-paclitaxel Squamous, any PD-L1 TPS 8.5 vs. 4.9, HR 0.37 (0.29 to 0.47) NR vs. 14.5, HR 0.55 (0.40 to 0.75) (143)
   GEMSTONE-302 Sugemalimab-platinum based chemotherapy Platinum based chemotherapy NSCLC, any PD-L1 TPS 9.0 vs. 4.9, HR 0.48 (0.39 to 0.60) 22.8 vs. 17.7, HR 0.67 (0.50 to 0.90) (144)
Anti-PD-(L)1 plus anti-CTLA4 +/- chemotherapy combination
   CheckMate 227 Nivolumab plus Ipilimumab Platinum based chemotherapy NSCLC, PD-L1
TPS ≥1%
5.1 vs. 5.6, HR 0.82 (0.69 to 0.97) 17.1 vs. 14.9, HR 0.79 (0.65 to 0.96) (145)
PD-L1 TPS <1% 5.1 vs. 4.7, HR 0.75 (o.59 to 0.96) 17.2 vs. 12.2, HR 0.62 (0.48 to 0.78)
   CheckMate 9LA Nivolumab, ipilimumab, platinum based chemotherapy Platinum based chemotherapy NSCLC, any
PD-L1 TPS
6.7 vs. 5.0, HR 0.68 (0.57 to 0.82) 15.6 vs. 10.9, HR 0.66 (0.55 to 0.80) (146)
   MYSTIC Durvalumab plus tremelimumab Platinum based chemotherapy NSCLC, PD-L1
TPS ≥25%
3.9 vs. 5.4, HR 1.05 (0.72 to 1.53) 11.9 vs. 12.9, HR 0.85 (0.61 to 1.17) (147)
   NEPTUNE Durvalumab plus tremelimumab Platinum based chemotherapy NSCLC with bTMB ≥20 mut/Mb 4.2 vs. 5.1, HR 0.77 (0.51 to 1.15) 11.7 vs. 9.1, HR 0.71 (0.49 to 1.05) (148)

NSCLC, non-small cell lung cancer; HR, hazard ratio; NR, not reached; OS, overall survival; PD-L1, programme death-ligand 1; PFS, progression-free survival; Ref, reference; TPS, tumor proportion score; bTMB, blood tumour mutational burden.

Addition of pembrolizumab to chemotherapy improved PFS and OS compared to chemotherapy alone regardless of PD-L1 expression and histology, and this regime has been adopted as the standard first-line treatment regime for advanced NSCLC without actionable alterations in multiple countries (131,133). Other studies testing immunochemotherapy combination with atezolizumab, sintilimab, tislelizumab, camrelizumab, or sugemalimab in addition to chemotherapy also consistently showed PFS improvement over chemotherapy alone (134-144) (Table 4). All of the studies above excluded patients with EGFR or ALK alterations. CheckMate 722 (NCT02864251) and KEYNOTE789 (NCT03515837) are phase III studies investigating the combination of anti-PD-(L)1 plus chemotherapy versus chemotherapy in advanced EGFR-mutated NSCLC NSCLC failing TKI. Results are eagerly awaited.

Despite the promise of anti-PD-(L)1 blockade, only a minority of patients experience durable benefit. Complex interplay between the immune system, tumour cells and tumour microenvironment (TME) are implied in the setting of immunotherapy resistance. ICI resistance can be broadly classified into tumour intrinsic and tumour extrinsic. Tumour intrinsic mechanisms include low tumour antigen production, defective antigen presentation or expression of co-inhibitory signals. Tumour extrinsic mechanisms include failure in T cell activation or infiltration into tumour, or intratumoral T cell dysfunction, either due to defects in host immune system or an immunosuppressive TME. To date, the landscape of ICI resistance is still poorly understood. Combinational strategies designed to overcome these resistance mechanisms are now investigated in clinical trials.

Immune checkpoints: CTLA4, TIGIT, and others

Cytotoxic T lymphocyte antigen 4 (CTLA4) is an immune checkpoint mechanistically distinct from PD-L1. It interferes with B7-CD28 binding and prevents T cell priming inside lymph nodes. It is also highly expressed by regulatory T (Treg) cells, thus blocking CTLA4 may both enhance T cell activation and downregulate Treg cells. CheckMate-227, MYSTIC and NEPTUNE study evaluated the anti-PD-L1/anti-CTLA4 combination without chemotherapy versus chemotherapy alone for treatment-naïve, EGFR/ALK negative, advanced NSCLC (Table 4). In the CheckMate-227 study, superior OS was achieved with nivolumab plus ipilimumab compared to chemotherapy, regardless of PD-L1 expression (146). Contrarily, both MYSTIC and NEPTUNE studies failed to achieve their primary endpoints—OS was not superior with dual immunotherapy in a biomarker-specified population (147,149). Both CheckMate-9LA and POSEIDON study showed that the four-drug combination of anti-PD-(L)1, anti-CTLA4 and chemotherapy was superior to chemotherapy alone in terms of PFS and OS, establishing the combination of dual immunotherapy plus chemotherapy as a valid first-line treatment option (146,150). Incidence of immune-related adverse events and rate of immunotherapy discontinuation was higher with dual immunotherapy compared to anti-PD-(L)1 monotherapy (145,149).

Prospective evidence is lacking in identifying patients who may benefit more from dual immunotherapy over anti-PD-(L)1 monotherapy. KEYNOTE-598 randomized patients with PD-L1 ≥50%, treatment-naïve advanced NSCLC to pembrolizumab single agent versus pembrolizumab plus ipilimumab. No PFS or OS improvement but only increased toxicities were observed in the dual immunotherapy arm (151). Based on this study, dual PD-(L)1 and CTLA blockade is not recommended for PD-L1 ≥50% NSCLCs.

Aside PD-L1 and CTLA4, multiple immune checkpoints are being evaluated as anticancer therapeutic targets. T cell immunoreceptor with Ig and ITIM domains (TIGIT) is a co-inhibitory immune receptor expressed on T cells. Preclinical data showed that dual TIGIT/PD-1 blockade enhanced antitumour immune response (152). The phase II CITYSCAPE trial randomly assigned 135 patients with treatment naïve, PD-L1 ≥1% to atezolizumab plus tiragolumab and atezolizumab alone. Patients in the atezolizumab plus tiragolumab arm achieved higher ORR (ORR 37% versus 21%), although the benefit was mainly driven by the PD-L1 ≥50% subgroup (ORR in PD-L1 ≥50% 69% versus 24%) (153). However, Roche recently reported that the phase III SKYSCRAPER-01 study, assessing tiragulomab plus atezolizumab versus atezolizumab alone for patients with PD-L1 high, EGFR/ALK negative, treatment naïve, advanced NSCLC, did not meet the co-primary end point of PFS (154). Apart from TIGIT, antibodies blocking other co-inhibitory immune checkpoints such as LAG3, TIM3 and VISTA and activating co-stimulatory receptors such as OX-40, ICOS, CD137 and GITR, are now studied in early-phase trials, either in monotherapy or in combination with anti-PD-(L)1 antibody (Table 5).

Table 5

Clinical Trials on novel targets to overcome ICI resistance in advanced NSCLC

Target Treatment strategy Setting Phase Study name (NCT identified)
Inhibitory immune checkpoints
   TIGIT Atezolizumab with or without tiragolumab Treatment naïve, high PD-L1 expression III SKYSCRAPER-01 (NCT04294810)
Pembrolizumab with or without vibostolimab Treatment naïve, PD-L1 TPS ≥1% III KEYVIBE-003 (NCT04738487)
Pembrolizumab plus chemotherapy with or without vibostolimab Treatment naive III KEYVIBE-007 (NCT05226598)
MK-7684A (pembrolizumab/vibostolimab coformulation) with or without docetaxel (vs. docetaxel) Prior ICI II KEYVIBE-002 (NCT04725188)
Pembrolizumab plus vibostolimab plus platinum based chemotherapy ICI naive I KEYVIBE-001 (NCT02964013)
Tislelizumab plus ociperilimab versus pembrolizumab or tislelizumab Treatment naïve, PD-L1 TPS ≥50% III (NCT04746924)
Tislelizumab plus chemotherapy with or without ociperlimab Treatment naive II (NCT05014815)
Zimberelimab (anti-PD-1) plus domvanalimab (anti-TIGIT) plus etrumadenant (A2R antagonist) Prior ICI, PD-L1 TPS ≥1% II (NCT04791839)
Zimberelimab or zimberelimab plus domvanalimab or zimberelimab plus domvanalimab plus etrumadenant Treatment naïve, PD-L1 TPS ≥50% II (NCT04262856)
Zimberelimab with or without domvanalimab, versus chemotherapy Treatment naïve, PD-L1 TPS ≥1% III (NCT04736173)
AZD2936 (anti-TIGIT/anti-PD-1 bispecific antibody) Prior ICI I/II (NCT04995523)
HLX301 (anti-TIGIT/anti-PD-1 bispecific antibody) Pretreated I/II (NCT05102214)
   LAG-3 XmAb22841 with or without pembrolizumab Pretreated I (NCT03849469)
RO7247669 monotherapy Pretreated I (NCT04140500)
Eftilagimoid alpha plus pembrolizumab Treatment naïve/prior ICI II (NCT03625323)
LAG525 with or without spartalizumab Pretreated I/II (NCT02460224)
   TIM-3 TSR-022 alone, or plus nivolumab or TSR-042 or TSR-033 or chemotherapy, or a combination of three drugs Pretreated I (NCT02817633)
RO7121661 monotherapy Pretreated I (NCT03708328)
INCAGNO2390 Pretreated I (NCT03652077)
BGB-A425 plus tislelizumab Pretreated I/II (NCT03744468)
AZD7789 (anti-PD-1/anti-TIM-3 bispecific antibody) Prior ICI I/II (NCT04931654)
MBG453 with or without spartalizumab Pretreated I/II (NCT02608268)
   S-15 NC318 monotherapy Pretreated I/II (NCT03665285)
NC318 plus pembrolizumab Prior ICI II (NCT04699123)
   VISTA HMBD-002 with or without pembrolizumab Pretreated I NCT05082610
Stimulatory signals
   OX-40 PF-04518600 with or without utomilumab Prior ICI I (NCT02315066)
INCAGN01949 plus nivolumab or ipilimumab or nivolumab-ipilimumab Pretreated I/II (NCT03241173)
SL-279252 (PD1-Fc-OX40L) Pretreated I (NCT03894618)
INBRX-106 with or without pembrolizumab Pretreated I (NCT04198766)
   CD40 APX005M plus nivolumab ICI naïve and pretreated I/II (NCT03123783)
CDX-1140, monotherapy, with CDX-301 (FLT3L), pembrolizumab or chemotherapy Pretreated I (NCT03329950)
SEA-CD40 with or without pembrolizumab Pretreated I (NCT02376699)
SEA-CD40 with pembrolizumab and platinum based chemotherapy ICI naive II (NCT04993677)
   ICOS Vopratelimab plus ipilimumab Prior ICI II (NCT03989362)
Vopratelimab plus JTX-4014 Pretreated II (NCT04549025)
GSK3359606 plus tremelimumab Pretreated I/II (NCT03693612)
GSK3359609 plus docetaxel Pretreated II (NCT03739710)
KY1044 with or without atezolizumab Pretreated I/II (NCT03829501)
   CD137 INBRX-105 with or without pembrolizumab Prior ICI I (NCT03809624)
   GITR INCAGN01876 plus nivolumab or ipilimumab or nivolumab-ipilimumab Pretreated I/II (NCT03126110)
VEGF
Pembrolizumab plus Lenvatinib or placebo Untreated, PD-L1 TPS ≥1% III LEAP-007 (NCT03829332)
Pembrolizumab plus platinum-pemetrexed plus lenvatinib or placebo Untreated, non squamous III LEAP-006 (NCT03829319)
Pembrolizumab plus Lenvatinib (versus docetaxel) Prior ICI and chemo III LEAP-008 (NCT03976375)
Nivolumab plus sitravatinib (versus docetaxel) Prior ICI and chemo III SAPPHIRE (NCT03906071)
Atezolizumab plus bevacizumab Untreated, TMB ≥10 II (NCT03836066)
Atezolizumab plus bevacizumab (versus atezolizumab) Untreated, PD-L1 TPS ≥1% II (NCT03896074)
Atezolizumab plus platinum-based chemotherapy plus bevacizumab Untreated II (NCT03713944)
Atezolizumab plus platinum-based chemotherapy with or without bevacizumab Untreated II (NCT03786692)
Atezolizumab plus ramucirumab Prior ICI II (NCT03689855)
Pembrolizumab plus ramucirumab (versus SOC) Prior ICI II (NCT03971474)
Pembrolizumab plus ramucirumab plus docetaxel Prior ICI and chemo II (NCT04340882)
Nivolumab plus ramucirumab Prior ICI II (NCT03527108)
Nivolumab plus ipilimumab plus nintedanib Treatment naïve and ICI pretreated I/II (NCT03377023)
Pembrolizumab plus lenvatinib Prior ICI I/II (NCT02501096)
Avelumab plus axitinib ICI naive II (NCT03472560)
Cancer vaccine
OSE2101 (vs. standard treatment) Prior ICI and chemo III ATALANTE 1 (NCT02654587)
RO7198457 with or without atezolizumab ICI naïve and pretreated I (NCT03289962)
Viagenpumatucel-L plus nivolumab or pembrolizumab plus pemetrexed Prior ICI I/II (NCT02439450)
NEO-PV-01 plus pembrolizumab plus platinum based chemotherapy Untreated I (NCT03380871)
GAd-PEV and MVA-PEV (personalized vaccine) with pembrolizumab ICI naïve, PD-L1 TPS ≥50% I (NCT04990479)
TG4010 (modified-human mucin 1-interleukin-2) vaccine with nivolumab ICI naive II (NCT02823990)
Adoptive cellular therapy
   TIL Autologous TILs plus nivolumab ICI naive I (NCT03215810)
LN-145 Prior ICI II (NCT04614103)
ATL001 with or without pembrolizumab Prior ICI I/II (NCT04032847)
ITIL306 Prior ICI and chemo I (NCT05397093)
   TCR T cells Anti-NY-ESO-1 TCR T cells Pretreated I/II (NCT05296564)
NY-ESO-1/LAGE-1a TCR T cells with or without pembrolizumab Pretreated II (NCT03709706)
Gene-edited autologous neoTCR-T cells monotherapy, or in combination with nivolumab or interleukin-2 Pretreated I (NCT03970382)
   CAR-T LYL797 (ROR1-targeted CAR T-cell therapy) Pretreated I (NCT05274451)
CAR-TnMUC1 Prior ICI and chemo I (NCT94925216)
   NK-cells GAIA-102 (activated NK-like cells) Pretreated I/II (NCT05207371)
Cytokine
   TGF-B Bintrafusp alfa (versus pembrolizumab) Untreated, PD-L1 TPS ≥50% III INTR@PID Lung 037 (NCT03631706)
Docetaxel with or without bintrafusp alfa Prior ICI and chemo II (NCT04396535)
TEW-7197 with durvalumab ICI naive I/II (NCT03732274)
   IL-1B Pembrolizumab plus platinum based chemotherapy plus canakinumab or placebo Untreated III CANOPY-1 (NCT03631199)
Docetaxel plus canakinumab or placebo Prior ICI and chemo III CANOPY-2 (NCT03626545)
Spartalizumab plus platinum based chemotherapy with or without canakinumab Prior ICI I (NCT03064854)
   IL-2 NKTR-214 plus nivolumab ICI naïve and pretreated I/II (NCT02983045)
THOR-707 with or without immune checkpoint inhibitor ICI naïve and pretreated I/II (NCT04009681)
Nemvaleukin alfa with or without pembrolizumab ICI naïve and pretreated I/II (NCT02799095)
STK-012 (pegylated engineered interleukin-2) Pretreated I (NCT05098132)
   IL-15 ALT-803 plus nivolumab Prior ICI I/II (NCT02523469)
ALT-803 plus immune checkpoint inhibitor Prior ICI II (NCT03228667)
   IL-12 Adenoviral-mediated interleukin (ADV/IL-12) gene therapy (intratumoral ADV/IL-12 injection) with atezolizumab Prior ICI I (NCT04911166)
Other TME targets
   IDO Pembrolizumab with or without epacadostat Untreated, PD-L1 TPS ≥50% II KEYNOTE-654-05 (NCT03322540)
Pembrolizumab plus platinum based chemotherapy with or without epacadostat Untreated II KEYNOTE-715-06 (NCT03322566)
Nivolumab plus ipilimumab plus epacadostat Pretreated I/II ECHO-208 (NCT03347123)
   HDAC Pembrolizumab plus mocetinostat plus guadecitabine Prior ICI I (NCT03220477)
Nivolumab plus glesatinib, sitravatinib, or mocetinostat Prior ICI II (NCT02954991)
Nivolumab plus entinostat plus azacytidine Prior chemo, ICI naïve or pretreated II (NCT01928576)
Pembrolizumab plus vorinostat ICI naïve and pretreated, PD-L1 TPS ≥1% I/II (NCT02638090)
Abexinostat plus pembrolizumab Pretreated I (NCT03590054)
   Adeno-sine Spartalizumab plus NIR178 Pretreated II (NCT03207867)
Durvalumab plus AZD4635 Prior ICI I (NCT02740985)
Etrumadenant plus zimberelimab Pretreated I (NCT03629756)
   CDK4/6 Pembrolizumab plus abemaciclib ICI naïve and pretreated Ib (NCT02779751)
   PARP Maintenance pembrolizumab plus olaparib vs. pembrolizumab plus pemetrexed Untreated, non-squamous maintenance III KEYLYNK-006 (NCT03976323)
Maintenance pembrolizumab plus olaparib or placebo Untreated, squamous, maintenance III KEYLYNK-008 (NCT03976362)
Avelumab plus talazoparib Prior ICI, STK11 mutation II (NCT04173507)
   MEK Pembrolizumab plus trametinib Pretreated, KRAS mutant I (NCT03299088)
Pembrolizumab plus trametinib pretreated I/II (NCT03225664)
Durvalumab plus tremelimumab plus selumetinib pretreated I/II (NCT03581487)
Pembrolizumab plus binimetinib Untreated, PD-L1 TPS ≥50% I (NCT03991819)
Atezolizumab plus cobimetinib Prior ICI II (NCT03600701)
   AXL Pembrolizumab plus bemcentinib ICI naïve and pretreated II (NCT03184571)

, not intended to be exhaustive. NSCLC, non-small cell lung cancer; NCT, National Clinical Trial; ICI, immune checkpoint inhibitor; PD-L1, programme death-ligand 1; TPS, tumour progression score.

Combination with anti-VEGF

Abnormal vasculature is a hallmark of cancer. Anti-angiogenesis agents can potentially modulate an immunosuppressive TME and enhance trafficking as well as tumour infiltration of immune cells (155).

Despite the proven synergism between anti-PD-(L)1 and anti-vascular endothelial growth factor (VEGF) in multiple cancer types, such evidence is more controversial in NSCLC (156-158). The IMpower150 study was a positive trial showing improved PFS and OS in patients who received atezolizumab-bevacizumab-chemotherapy compared to bevacizumab-chemotherapy, but this study did not directly answer the question whether bevacizumab had added value on top of PD-L1 blockade (134). LEAP-007 randomized patients with untreated, EGFR/ALK negative, PD-L1 TPS ≥1% advanced NSCLC to pembrolizumab versus pembrolizumab plus lenvatinib. This was a negative study (OS 14.1 versus 16.4 months, HR 1.10, OS numerically lower in pembrolizumab/lenvatinib arm) (159). However, Reckamp et al. recently reported in a phase II study in which patients who were previously treated with ICI and platinum-based chemotherapy, OS was significantly improved with pembrolizumab plus ramucirumab compared to chemotherapy of physician’s choice [OS 14.5 versus 11.6 months, HR 0.69 (80% CI: 0.51–0.92)] (160). This suggested that antiangiogenic agents did resensitize tumours to ICI in a certain subset of patients. Ongoing clinical studies with biomarker analyses will further define patients who may potentially benefit from antiangiogenic agents (Table 5).

Cancer vaccines

Low tumour neoantigen expression and defective antigen presentation are two fundamental reasons of impaired T cell activation and ICI failure. Cancer vaccine primarily works by enhancing antigen-specific T cell antitumour responses. Most cancer vaccines tested in lung cancer clinical trials utilized tumour-associated antigens (TAAs), such as melanoma-associated antigen-A3 (MAGE-A3) and mucin 1 (MUC-1), self-antigens that are commonly overexpressed but not uniquely expressed on tumour cells. Two phase III studies, MAGRIT and START, evaluated MAGE-A3 vaccine and MUG1 antigen vaccine versus placebo respectively in the adjuvant setting. Both studies failed to report survival benefit with the study treatment (161,162).

Following the disappointing results of single-antigen vaccination, clinical research now focuses on multiple-antigen vaccination and personalized antigen vaccination. In the first part of the phase III ATALANTE-1 randomised trial, OSE-2101, a HLA-A2 restricted neoepitope vaccine targeting five TAAs (CEA, HER2, MAGE2, MAGE3, and P53) expressed on lung cancer cells, achieved superior OS over standard-of-care second-line chemotherapy in patients failing platinum-based chemotherapy and anti-PD-(L)1 treatment (163). Part 2 is now ongoing and will enroll 363 participants with OS as the primary endpoint (NCT02654587). Personalized antigen vaccination is constructed by whole exome and RNA sequencing of the tumour and identification of personalized immunogenic neoepitopes based on bioinformatic algorithms (164). In a phase Ib study, response rate was 39% with no extra safety signals reported in 18 patients in the NSCLC cohort who received NEO-PV-01 (a personalized neoantigen vaccine) concurrently with nivolumab (165). Multiple studies on personalized cancer vaccine are now ongoing either as monotherapy or given in combination with ICIs (Table 5).

Adoptive cellular therapy

Given the vital role of T cell in antitumour immunity, adoptive cellular therapy (ACT) developed to date has mainly focused on T cell therapy, although other cellular therapies (e.g., NK cell, B cell, macrophage) have demonstrated therapeutic potentials as well. Similar to cancer vaccine, the objective of ACT is to generate tumour-specific, activated immune cells (166). Adoptive T cell therapy includes tumour-infiltrating lymphocyte (TIL) therapy, T-cell receptor (TCR)-engineered T cell therapy and chimeric antigen receptor T-cell (CAR-T) therapy. TILs are polyclonal T cells isolated from patient’s tumour tissue, expanded ex-vivo and reinfused into the patient after lymphodepletion. In a recent phase I trial, TIL therapy was administered with nivolumab in 20 patients with advanced NSCLC upon progression on anti-PD-(L)1 treatment. The study met the end point of safety. Objective response was noted in three out of thirteen evaluable patients including two with durable complete responses (167). In TCR-engineered T cell and CAR-T cell therapy, autologous T cells are isolated from peripheral circulation followed by ex-vivo transduction of a tumour-antigen specific TCR or CAR (166). Despite its tremendous success in treating haematological malignancies, CAR-T for solid tumour treatment proves to be challenging due to factors like poor tumour infiltration, immunosuppressive TME, and off-target toxicities (168). Innovative approaches have been evaluated to overcome these resistance mechanisms. For instance, gene-edited T cells may enhance both safety as well as antitumour efficacy. In a phase I first-in-human study, CRISPR-Cas9 PD-1-edited T cells were safely given to 12 patients with advanced NSCLC without severe toxicities (169). Combination of cellular therapy with other modalities of immunotherapy, such as PD-L1 blockade or cytokine therapy, may further potentiate the therapeutic efficacy of tumour-specific immune cells (170,171).

Targeting tumour microenvironment, cytokine, and gut microbiota

Cytokines are pivotal in modulating immune cell function and thus have been actively investigated in enhancing antitumour efficacy of immunotherapy. This concept is supported by the fact that a minority of patients with melanoma and renal cell carcinoma achieved durable responses with high dose interleukin-2 (IL-2) therapy (172,173). A phase I study showed that the combination of NKTR-214 (a prodrug of polyethylene glycol (PEG)-conjugated IL-2) and nivolumab was safe and efficacious (three out of five patients with NSCLC achieved RECIST response) in immunotherapy-naïve patients with various cancer types including NSCLC (174,175). A global dose escalation study for treatment-naïve lung cancer patients is ongoing (NCT03138889). Another phase I study evaluated the combination of nivolumab and ALT-803, a modified IL-15/IL-15Rα/IgG1 Fc complex in patients with anti-PD-L1 antibody refractory NSCLC (NCT02523469). Twenty-one patients received study treatment and ORR was 29% (176).

Canakinumab, an anti-IL-1B monoclonal antibody, has been studied in multiple phase II and III studies after a significant reduction in lung cancer incidence and mortality was observed in the large scale CANTOS study which investigated the role of IL-1B in atherosclerosis (177). However, both the phase III CANOPY-1 (pembrolizumab plus chemotherapy with or without canakinumab in untreated metastatic NSCLC, NCT03631199) and CANOPY-2 (docetaxel with or without canakinumab in pretreated NSCLC) studies failed to improve PFS and OS (178,179). CANOPY-A and CANOPY-N are ongoing studies evaluating the drug in the neoadjuvant and adjuvant setting in early staged disease (NCT03447769, NCT03968419).

A multitude of enzymatic pathways and cellular components are associated with an immunosuppressive TME. These molecules have been explored as therapeutic targets in hope to modulate the TME and enhance immune activity. Histone deacetylase (HDAC) alters gene expression via epigenetic regulation and HDAC inhibitors are found to promote pro-immune activity within the TME. Two phase II studies reported antitumour activity and acceptable safety profile of HDAC inhibitor and anti-PD-(L)1 combination in patients with PD-(L)1 resistant/refractory NSCLC (180,181). Further clinical studies are warranted to select patients who may benefit from this drug combination. Other targets of interest, including indoleamine 2,3-dioxygenase (IDO), RORγ, AXL, MEK, PARP and CD73, are now being studied in early phase clinical trials enrolling NSCLC patients (Table 5).

Lastly, gut microbiota has emerged as a potential immune mediator that may affect clinical response to ICI treatment. Preclinical studies showed that a favourable gut flora composition is predictive of ICI response and modulation of gut flora may reverse ICI resistance (182). This strategy is now tested in prospective trials for patients with ICI failure, mainly by introducing favourable gut flora via faecal microbial transplantation and ICI rechallenge.

Immunotherapy in advanced NSCLC: future direction

Although the adoption of PD-L1 blockade has marked a new milestone of NSCLC management, ICI resistance and lack of predictive biomarker remain two major challenges. While various resistance models have been proposed, ICI resistance in reality is likely multifactorial and methods for ascertaining ICI resistance mechanisms are currently lacking. Combinational immunotherapy approaches targeting different phases of antitumour immunity have shown encouraging signals. However, most of these approaches were not tested in a biomarker-based fashion, which might explain the failure of some phase II/III trials despite favourable results seen in early phase studies. More translational research is required to identify predictive biomarkers of ICI response or resistance. Advances in diagnostic and analytic tools like multiomic analysis, single cell sequencing, and deep learning approaches, may offer further insight into the dynamic interaction between the host immune system, cancer cells and TME.


Strengths and limitations of this review

This review offers a succinct overview in the latest update in targeted therapy and immunotherapy for NSCLC management, identified resistance mechanisms, and current research direction to overcome treatment resistance and further improve treatment outcome. Owing to the breadth of the topic, some sections cannot be discussed in depth. While the authors agree that the advances in molecular diagnostics and biomarker research is also crucial for the successful development of novel therapeutics, this topic is beyond the scope of this review.


Conclusions

Deeper understanding of the molecular and immune biology of NSCLC has led to rapid development of targeted therapy and immunotherapy in this field. The indications of targeted therapy and immunotherapy have now extended to early staged NSCLC as randomized studies have confirmed that these agents reduce disease recurrence. Comprehensive genomic profiling at diagnosis of advanced disease maximizes the chance for patient to benefit from highly efficacious molecular therapies. At the time of treatment resistance, next generation molecular therapies and novel drug combinations tailored to specific resistance mechanisms may now be considered. Currently, only a minority of patients experience durable clinical benefit with immunotherapy and innovative strategies targeting various immune resistance mechanisms have yielded early promise. As the therapeutic landscape of NSCLC is becoming increasingly complex, identification of predictive and resistance biomarkers is key to success in future drug development. The importance of multiomic profiling and translational studies alongside clinical trials cannot be overemphasized.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned by the Guest Editor (Calvin S. H. Ng) for the series “Lung Cancer Management—The Next Decade” published in Annals of Translational Medicine. The article has undergone external peer review.

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://atm.amegroups.com/article/view/10.21037/atm-22-4444/rc

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://atm.amegroups.com/article/view/10.21037/atm-22-4444/coif). The series “Lung Cancer Management—The Next Decade” was commissioned by the editorial office without any funding or sponsorship. MSCL reports honoraria as a speaker for Novartis and advisory fees from Pfizer. KKSM reports reimbursement for travel expenses for ESMO course. TSKM reports research grants (paid to institution) from AstraZeneca, Bristol Myers Squibb, Pfizer, Roche, Novartis, SFJ Pharmaceuticals, MSD, G1 Therapeutics, Takeda, XCovery and Merck Serono; consulting fees from Amoy Diagnostics, Boehringer Ingelheim, Amgen, Bristol Myers Squibb, C4 Therapeutics, Curio Science, Daiichi Sankyo, Eisai, Gilead Sciences, Guardant Health, Ignyta, Inivata, Janssen, Lunit USA, Lucence Health, MSD, MoreHealth, Origimed, Pfizer, Qiming Development (HK), Sanofi-Aventis, Takeda Pharmaceuticals, Yuhan Corporation, Berry Oncology, AstraZeneca, BeiGene, Blueprint Medicines, Alpha Biopharma, Covidien LP, CStone Pharmaceuticals, D3 Bio Ltd., Fishawack Facilitate, Elevation Oncology, Gritstone Oncology, Hengrui Therapeutics, Incyte Corporation, IQVIA, Lilly, Loxo-Oncology, Merck Serono, Mirati Therapeutics, Novartis, OSE Immunotherapeutics, Puma Biotechnology, Roche Pharmaceuticals, SFJ Pharmaceutical and Vertex Pharmaceuticals; speaker fees from ACEA Pharmaceuticals, Amgen, AstraZeneca, Boehringer Ingelheim Pharmaceuticals Inc., Daiichi Sankyo, Fishawack Facilitate, Janssen, Lucence Health, Medscape LLC, Merck Sharp & Dohme, OrigiMed Co., Ltd., PeerVoice, Pfizer, Research to Practice, Sanofi-Aventis, Liangyihui Network Technology, Takeda Oncology, Alpha Biopharma, Amoy Diagnostics, BeiGene, Bristol Myers Squibb, Daz Group, InMed Medical Communication, Lilly, MD Health Brazil, Merck, Novartis, P. Permanyer SL, Physicians’ Education Resource, PrIME Oncology, Roche Pharmaceuticals/Diagnostics/Foundation One, Shanghai BeBirds Translation & Consulting, Taiho Pharmaceutical and Touch Independent Medical Education; he serves as the Advisory Board Member for AbbVie, Amgen, Berry Oncology, Boehringer Ingelheim Pharmaceuticals, C4 Therapeutics, CStone Pharmaceuticals, D3 Bio Ltd., Eisai, G1 Therapeutics Inc., Gritstone Oncology Inc., GeneDecode Co Ltd, Hutchmed, Incyte Corporation, IQVIA, Lilly, Lunit, Merck Sharp & Dohme, Novartis, Puma Biotechnology, Roche/Genentech, SFJ Pharmaceutical, Vertex Pharmaceuticals, Yuhan Corporation, ACEA Pharma, AstraZeneca, Blueprint Medicines Corporation, Bristol-Myers Squibb Company, Covidien LP, Curio Science, Daiichi Sankyo, Fishawack Facilitate, Gilead Sciences, Guardant Health, Hengrui Therapeutics, Ignyta, Inivata, Janssen, Loxo-Oncology, Merck Serono, Mirati Therapeutics, Origimed, Pfizer, Sanofi-Aventis R&D, Takeda and Virtus Medical Group; he serves as the Board of Directors (remunerated) for AstraZeneca PLC, Lunit USA, HutchMed and ACT Genomics-Sanomics Group; he has the leadership role (non-remunerated) in Chinese Society of Clinical Oncology (CSCO), Hong Kong Cancer Therapy Society (HKCTS), Asian Thoracic Oncology Research Group (ATORG), American Society of Clinical Oncology (ASCO), Chinese Lung Cancer Research Foundation Limited (CLCRF), Hong Kong Cancer Fund (HKCF) and International Association for the Study of Lung Cancer (IASLC). Besides, he holds equities in Aurora Tele-Oncology Ltd., HutchMed and ACT Genomics-Sanomics Group. The authors have no other conflicts of interest to declare.

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

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


References

  1. Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin 2021;71:209-49. [Crossref] [PubMed]
  2. Woodard GA, Jones KD, Jablons DM. Lung Cancer Staging and Prognosis. Cancer Treat Res 2016;170:47-75. [Crossref] [PubMed]
  3. Scagliotti GV, Parikh P, von Pawel J, et al. Phase III study comparing cisplatin plus gemcitabine with cisplatin plus pemetrexed in chemotherapy-naive patients with advanced-stage non-small-cell lung cancer. J Clin Oncol 2008;26:3543-51. [Crossref] [PubMed]
  4. Preoperative chemotherapy for non-small-cell lung cancer: a systematic review and meta-analysis of individual participant data. Lancet 2014;383:1561-71. [Crossref] [PubMed]
  5. Pignon JP, Tribodet H, Scagliotti GV, et al. Lung adjuvant cisplatin evaluation: a pooled analysis by the LACE Collaborative Group. J Clin Oncol 2008;26:3552-9. [Crossref] [PubMed]
  6. Doroshow DB, Sanmamed MF, Hastings K, et al. Immunotherapy in Non-Small Cell Lung Cancer: Facts and Hopes. Clin Cancer Res 2019;25:4592-602. [Crossref] [PubMed]
  7. Forde PM, Chaft JE, Smith KN, et al. Neoadjuvant PD-1 Blockade in Resectable Lung Cancer. N Engl J Med 2018;378:1976-86. [Crossref] [PubMed]
  8. Pataer A, Kalhor N, Correa AM, et al. Histopathologic response criteria predict survival of patients with resected lung cancer after neoadjuvant chemotherapy. J Thorac Oncol 2012;7:825-32. [Crossref] [PubMed]
  9. William WN Jr, Pataer A, Kalhor N, et al. Computed tomography RECIST assessment of histopathologic response and prediction of survival in patients with resectable non-small-cell lung cancer after neoadjuvant chemotherapy. J Thorac Oncol 2013;8:222-8. [Crossref] [PubMed]
  10. Carbone D, Lee J, Kris M, et al. OA06.06 Clinical/Biomarker Data for Neoadjuvant Atezolizumab in Resectable Stage IB-IIIB NSCLC: Primary Analysis in the LCMC3 Study. J Thorac Oncol 2021;16:S115-6. [Crossref]
  11. Gao S, Li N, Gao S, et al. Neoadjuvant PD-1 inhibitor (Sintilimab) in NSCLC. J Thorac Oncol 2020;15:816-26. [Crossref] [PubMed]
  12. Cascone T, William WN Jr, Weissferdt A, et al. Neoadjuvant nivolumab or nivolumab plus ipilimumab in operable non-small cell lung cancer: the phase 2 randomized NEOSTAR trial. Nat Med 2021;27:504-14. [Crossref] [PubMed]
  13. Provencio M, Nadal E, Insa A, et al. Neoadjuvant chemotherapy and nivolumab in resectable non-small-cell lung cancer (NADIM): an open-label, multicentre, single-arm, phase 2 trial. Lancet Oncol 2020;21:1413-22. [Crossref] [PubMed]
  14. Shu CA, Gainor JF, Awad MM, et al. Neoadjuvant atezolizumab and chemotherapy in patients with resectable non-small-cell lung cancer: an open-label, multicentre, single-arm, phase 2 trial. Lancet Oncol 2020;21:786-95. [Crossref] [PubMed]
  15. Rothschild SI, Zippelius A, Eboulet EI, et al. SAKK 16/14: Durvalumab in Addition to Neoadjuvant Chemotherapy in Patients With Stage IIIA(N2) Non–Small-Cell Lung Cancer—A Multicenter Single-Arm Phase II Trial. J Clin Oncol 2021;39:2872-80. [Crossref] [PubMed]
  16. Forde PM, Spicer J, Lu S, et al. Neoadjuvant Nivolumab plus Chemotherapy in Resectable Lung Cancer. N Engl J Med 2022;386:1973-85. [Crossref] [PubMed]
  17. Felip E, Altorki N, Zhou C, et al. Adjuvant atezolizumab after adjuvant chemotherapy in resected stage IB-IIIA non-small-cell lung cancer (IMpower010): a randomised, multicentre, open-label, phase 3 trial. Lancet 2021;398:1344-57. [Crossref] [PubMed]
  18. O'Brien M, Paz-Ares L, Marreaud S, et al. Pembrolizumab versus placebo as adjuvant therapy for completely resected stage IB-IIIA non-small-cell lung cancer (PEARLS/KEYNOTE-091): an interim analysis of a randomised, triple-blind, phase 3 trial. Lancet Oncol 2022;23:1274-86. [Crossref] [PubMed]
  19. Antonia SJ, Villegas A, Daniel D, et al. Durvalumab after Chemoradiotherapy in Stage III Non-Small-Cell Lung Cancer. N Engl J Med 2017;377:1919-29. [Crossref] [PubMed]
  20. Spigel DR, Faivre-Finn C, Gray JE, et al. Five-Year Survival Outcomes From the PACIFIC Trial: Durvalumab After Chemoradiotherapy in Stage III Non-Small-Cell Lung Cancer. J Clin Oncol 2022;40:1301-11. [Crossref] [PubMed]
  21. Herbst RS, Majem M, Barlesi F, et al. COAST: An Open-Label, Phase II, Multidrug Platform Study of Durvalumab Alone or in Combination With Oleclumab or Monalizumab in Patients With Unresectable, Stage III Non-Small-Cell Lung Cancer. J Clin Oncol 2022;40:3383-93. [Crossref] [PubMed]
  22. Jabbour SK, Lee KH, Frost N, et al. Pembrolizumab Plus Concurrent Chemoradiation Therapy in Patients With Unresectable, Locally Advanced, Stage III Non-Small Cell Lung Cancer: The Phase 2 KEYNOTE-799 Nonrandomized Trial. JAMA Oncol 2021; Epub ahead of print. [Crossref] [PubMed]
  23. Wu YL, Zhong W, Chen KN, et al. CTONG1103: Final overall survival analysis of the randomized phase 2 trial of erlotinib versus gemcitabine plus cisplatin as neoadjuvant treatment of stage IIIA-N2 EGFR-mutant non–small cell lung cancer. J Clin Oncol 2021;39:8502. [Crossref]
  24. Lyu C, Fang W, Ma H, et al. Osimertinib as neoadjuvant treatment for resectable stage II-IIIB EGFR mutant lung adenocarcinoma (NEOS). J Clin Oncol 2021;39:8524. [Crossref]
  25. Wu YL, Tsuboi M, He J, et al. Osimertinib in Resected EGFR-Mutated Non-Small-Cell Lung Cancer. N Engl J Med 2020;383:1711-23. [Crossref] [PubMed]
  26. Sholl LM, Aisner DL, Varella-Garcia M, et al. Multi-institutional Oncogenic Driver Mutation Analysis in Lung Adenocarcinoma: The Lung Cancer Mutation Consortium Experience. J Thorac Oncol 2015;10:768-77. [Crossref] [PubMed]
  27. Gou LY, Wu YL. Prevalence of driver mutations in non-small-cell lung cancers in the People's Republic of China. Lung Cancer (Auckl) 2014;5:1-9. [PubMed]
  28. Jordan EJ, Kim HR, Arcila ME, et al. Prospective Comprehensive Molecular Characterization of Lung Adenocarcinomas for Efficient Patient Matching to Approved and Emerging Therapies. Cancer Discov 2017;7:596-609. [Crossref] [PubMed]
  29. Mok TS, Wu YL, Thongprasert S, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med 2009;361:947-57. [Crossref] [PubMed]
  30. Fukuoka M, Wu YL, Thongprasert S, et al. Biomarker analyses and final overall survival results from a phase III, randomized, open-label, first-line study of gefitinib versus carboplatin/paclitaxel in clinically selected patients with advanced non-small-cell lung cancer in Asia (IPASS). J Clin Oncol 2011;29:2866-74. [Crossref] [PubMed]
  31. Maemondo M, Inoue A, Kobayashi K, et al. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med 2010;362:2380-8. [Crossref] [PubMed]
  32. Inoue A, Kobayashi K, Maemondo M, et al. Updated overall survival results from a randomized phase III trial comparing gefitinib with carboplatin-paclitaxel for chemo-naïve non-small cell lung cancer with sensitive EGFR gene mutations (NEJ002). Ann Oncol 2013;24:54-9. [Crossref] [PubMed]
  33. Rosell R, Carcereny E, Gervais R, et al. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol 2012;13:239-46. [Crossref] [PubMed]
  34. Zhou C, Wu YL, Chen G, et al. Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol 2011;12:735-42. [Crossref] [PubMed]
  35. Zhou C, Wu YL, Chen G, et al. Final overall survival results from a randomised, phase III study of erlotinib versus chemotherapy as first-line treatment of EGFR mutation-positive advanced non-small-cell lung cancer (OPTIMAL, CTONG-0802). Ann Oncol 2015;26:1877-83. [Crossref] [PubMed]
  36. Park K, Tan EH, O'Byrne K, et al. Afatinib versus gefitinib as first-line treatment of patients with EGFR mutation-positive non-small-cell lung cancer (LUX-Lung 7): a phase 2B, open-label, randomised controlled trial. Lancet Oncol 2016;17:577-89. [Crossref] [PubMed]
  37. Paz-Ares L, Tan EH, O'Byrne K, et al. Afatinib versus gefitinib in patients with EGFR mutation-positive advanced non-small-cell lung cancer: overall survival data from the phase IIb LUX-Lung 7 trial. Ann Oncol 2017;28:270-7. [Crossref] [PubMed]
  38. Wu YL, Cheng Y, Zhou X, et al. Dacomitinib versus gefitinib as first-line treatment for patients with EGFR-mutation-positive non-small-cell lung cancer (ARCHER 1050): a randomised, open-label, phase 3 trial. Lancet Oncol 2017;18:1454-66. [Crossref] [PubMed]
  39. Mok TS, Cheng Y, Zhou X, et al. Improvement in Overall Survival in a Randomized Study That Compared Dacomitinib With Gefitinib in Patients With Advanced Non-Small-Cell Lung Cancer and EGFR-Activating Mutations. J Clin Oncol 2018;36:2244-50. [Crossref] [PubMed]
  40. Mok TS, Wu Y-L, Ahn M-J, et al. Osimertinib or Platinum-Pemetrexed in EGFR T790M-Positive Lung Cancer. N Engl J Med 2017;376:629-40. [Crossref] [PubMed]
  41. Papadimitrakopoulou VA, Mok TS, Han JY, et al. Osimertinib versus platinum-pemetrexed for patients with EGFR T790M advanced NSCLC and progression on a prior EGFR-tyrosine kinase inhibitor: AURA3 overall survival analysis. Ann Oncol 2020;31:1536-44. [Crossref] [PubMed]
  42. Soria JC, Ohe Y, Vansteenkiste J, et al. Osimertinib in Untreated EGFR-Mutated Advanced Non-Small-Cell Lung Cancer. N Engl J Med 2018;378:113-25. [Crossref] [PubMed]
  43. Ramalingam SS, Vansteenkiste J, Planchard D, et al. Overall Survival with Osimertinib in Untreated, EGFR-Mutated Advanced NSCLC. N Engl J Med 2020;382:41-50. [Crossref] [PubMed]
  44. Solomon BJ, Mok T, Kim DW, et al. First-line crizotinib versus chemotherapy in ALK-positive lung cancer. N Engl J Med 2014;371:2167-77. [Crossref] [PubMed]
  45. Solomon BJ, Kim DW, Wu YL, et al. Final Overall Survival Analysis From a Study Comparing First-Line Crizotinib Versus Chemotherapy in ALK-Mutation-Positive Non-Small-Cell Lung Cancer. J Clin Oncol 2018;36:2251-8. [Crossref] [PubMed]
  46. Soria JC, Tan DSW, Chiari R, et al. First-line ceritinib versus platinum-based chemotherapy in advanced ALK-rearranged non-small-cell lung cancer (ASCEND-4): a randomised, open-label, phase 3 study. Lancet 2017;389:917-29. [Crossref] [PubMed]
  47. Mok T, Camidge DR, Gadgeel SM, et al. Updated overall survival and final progression-free survival data for patients with treatment-naive advanced ALK-positive non-small-cell lung cancer in the ALEX study. Ann Oncol 2020;31:1056-64. [Crossref] [PubMed]
  48. Camidge DR, Kim HR, Ahn MJ, et al. Brigatinib Versus Crizotinib in ALK Inhibitor-Naive Advanced ALK-Positive NSCLC: Final Results of Phase 3 ALTA-1L Trial. J Thorac Oncol 2021;16:2091-108. [Crossref] [PubMed]
  49. Horn L, Infante JR, Reckamp KL, et al. Ensartinib (X-396) in ALK-Positive Non-Small Cell Lung Cancer: Results from a First-in-Human Phase I/II, Multicenter Study. Clin Cancer Res 2018;24:2771-9. [Crossref] [PubMed]
  50. Shaw AT, Bauer TM, de Marinis F, et al. First-Line Lorlatinib or Crizotinib in Advanced ALK-Positive Lung Cancer. N Engl J Med 2020;383:2018-29. [Crossref] [PubMed]
  51. Leonetti A, Sharma S, Minari R, et al. Resistance mechanisms to osimertinib in EGFR-mutated non-small cell lung cancer. Br J Cancer 2019;121:725-37. [Crossref] [PubMed]
  52. Schalm SS, Dineen T, Lim SM, et al. 1296P BLU-945, a highly potent and selective 4th generation EGFR TKI for the treatment of EGFR T790M/C797S resistant NSCLC. Ann Oncol 2020;31:S839. [Crossref]
  53. Conti C, Campbell J, Woessner R, et al. Abstract 1262: BLU-701 is a highly potent, brain-penetrant and WT-sparing next-generation EGFR TKI for the treatment of sensitizing (ex19del, L858R) and C797S resistance mutations in metastatic NSCLC. Cancer Res 2021;81:1262. [Crossref]
  54. To C, Beyett TS, Jang J, et al. An allosteric inhibitor against the therapy-resistant mutant forms of EGFR in non-small cell lung cancer. Nat Cancer 2022;3:402-17. [Crossref] [PubMed]
  55. Lim SM, Kim DW, Jung JE, et al. 1365TiP A phase I/II, open-label study of BBT-176, a triple mutation targeting EGFR TKI, in patients with NSCLC who progressed after prior EGFR TKI therapy. Ann Oncol 2021;32:S1035. [Crossref]
  56. Sequist LV, Han JY, Ahn MJ, et al. Osimertinib plus savolitinib in patients with EGFR mutation-positive, MET-amplified, non-small-cell lung cancer after progression on EGFR tyrosine kinase inhibitors: interim results from a multicentre, open-label, phase 1b study. Lancet Oncol 2020;21:373-86. [Crossref] [PubMed]
  57. Wu YL, Cheng Y, Zhou J, et al. Tepotinib plus gefitinib in patients with EGFR-mutant non-small-cell lung cancer with MET overexpression or MET amplification and acquired resistance to previous EGFR inhibitor (INSIGHT study): an open-label, phase 1b/2, multicentre, randomised trial. Lancet Respir Med 2020;8:1132-43. [Crossref] [PubMed]
  58. Wu YL, Zhang L, Kim DW, et al. Phase Ib/II Study of Capmatinib (INC280) Plus Gefitinib After Failure of Epidermal Growth Factor Receptor (EGFR) Inhibitor Therapy in Patients With EGFR-Mutated, MET Factor-Dysregulated Non-Small-Cell Lung Cancer. J Clin Oncol 2018;36:3101-9. [Crossref] [PubMed]
  59. McCoach CE, Yu A, Gandara DR, et al. Phase I/II Study of Capmatinib Plus Erlotinib in Patients With MET-Positive Non-Small-Cell Lung Cancer. JCO Precis Oncol 2021;1:PO.20.00279.
  60. Rehman M, Kim C, Reuss JE, et al. Divergent RET- and BRAF-Mediated Resistance to Osimertinib in EGFR-Mutant NSCLC: A Case Report. JCO Precis Oncol 2021;5:939-42. [Crossref] [PubMed]
  61. Hou H, Sun D, Zhang C, et al. ALK rearrangements as mechanisms of acquired resistance to osimertinib in EGFR mutant non-small cell lung cancer. Thorac Cancer 2021;12:962-9. [Crossref] [PubMed]
  62. Zhu Y, Zhang Q, Wang W, et al. P88.02 SDC4-ROS1 Fusion as a Mechanism of Acquired Resistance in EGFR-Mutant Lung Adenocarcinoma. J Thorac Oncol 2021;16:S686-7. [Crossref]
  63. Syed YY. Amivantamab: First Approval. Drugs 2021;81:1349-53. [Crossref] [PubMed]
  64. Leighl NB, Shu CA, Minchom A, et al. 1192MO Amivantamab monotherapy and in combination with lazertinib in post-osimertinib EGFR-mutant NSCLC: Analysis from the CHRYSALIS study. Ann Oncol 2021;32:S951-2. [Crossref]
  65. Shu CA, Goto K, Ohe Y, et al. 1193MO Amivantamab plus lazertinib in post-osimertinib, post-platinum chemotherapy EGFR-mutant non-small cell lung cancer (NSCLC): Preliminary results from CHRYSALIS-2. Ann Oncol 2021;32:S952-3. [Crossref]
  66. Moores SL, Chiu ML, Bushey BS, et al. A Novel Bispecific Antibody Targeting EGFR and cMet Is Effective against EGFR Inhibitor-Resistant Lung Tumors. Cancer Res 2016;76:3942-53. [Crossref] [PubMed]
  67. Kawano O, Sasaki H, Endo K, et al. ErbB3 mRNA expression correlated with specific clinicopathologic features of Japanese lung cancers. J Surg Res 2008;146:43-8. [Crossref] [PubMed]
  68. Jänne PA, Baik C, Su WC, et al. Efficacy and Safety of Patritumab Deruxtecan (HER3-DXd) in EGFR Inhibitor-Resistant, EGFR-Mutated Non-Small Cell Lung Cancer. Cancer Discov 2022;12:74-89. [Crossref] [PubMed]
  69. Peters S, Camidge DR, Shaw AT, et al. Alectinib versus Crizotinib in Untreated ALK-Positive Non-Small-Cell Lung Cancer. N Engl J Med 2017;377:829-38. [Crossref] [PubMed]
  70. Solomon B, Bauer T, Mok T, et al. Abstract CT223: Updated efficacy and safety from the phase 3 CROWN study of first-line lorlatinib vs. crizotinib in advanced anaplastic lymphoma kinase (ALK)-positive non-small cell lung cancer (NSCLC). Cancer Res 2022;82:CT223. [Crossref]
  71. Solomon BJ, Besse B, Bauer TM, et al. Lorlatinib in patients with ALK-positive non-small-cell lung cancer: results from a global phase 2 study. Lancet Oncol 2018;19:1654-67. [Crossref] [PubMed]
  72. Gainor JF, Dardaei L, Yoda S, et al. Molecular Mechanisms of Resistance to First- and Second-Generation ALK Inhibitors in ALK-Rearranged Lung Cancer. Cancer Discov 2016;6:1118-33. [Crossref] [PubMed]
  73. Yoda S, Lin JJ, Lawrence MS, et al. Sequential ALK Inhibitors Can Select for Lorlatinib-Resistant Compound ALK Mutations in ALK-Positive Lung Cancer. Cancer Discov 2018;8:714-29. [Crossref] [PubMed]
  74. Murray BW, Zhai D, Deng W, et al. TPX-0131, a Potent CNS-penetrant, Next-generation Inhibitor of Wild-type ALK and ALK-resistant Mutations. Mol Cancer Ther 2021;20:1499-507. [Crossref] [PubMed]
  75. Shaw AT, Ou SH, Bang YJ, et al. Crizotinib in ROS1-rearranged non-small-cell lung cancer. N Engl J Med 2014;371:1963-71. [Crossref] [PubMed]
  76. Drilon A, Siena S, Dziadziuszko R, et al. Entrectinib in ROS1 fusion-positive non-small-cell lung cancer: integrated analysis of three phase 1-2 trials. Lancet Oncol 2020;21:261-70. [Crossref] [PubMed]
  77. Dziadziuszko R, Krebs MG, De Braud F, et al. Updated Integrated Analysis of the Efficacy and Safety of Entrectinib in Locally Advanced or Metastatic ROS1 Fusion-Positive Non-Small-Cell Lung Cancer. J Clin Oncol 2021;39:1253-63. [Crossref] [PubMed]
  78. Lim SM, Kim HR, Lee JS, et al. Open-Label, Multicenter, Phase II Study of Ceritinib in Patients With Non-Small-Cell Lung Cancer Harboring ROS1 Rearrangement. J Clin Oncol 2017;35:2613-8. [Crossref] [PubMed]
  79. Shaw AT, Solomon BJ, Chiari R, et al. Lorlatinib in advanced ROS1-positive non-small-cell lung cancer: a multicentre, open-label, single-arm, phase 1-2 trial. Lancet Oncol 2019;20:1691-701. [Crossref] [PubMed]
  80. Lin JJ, Choudhury NJ, Yoda S, et al. Spectrum of Mechanisms of Resistance to Crizotinib and Lorlatinib in ROS1 Fusion-Positive Lung Cancer. Clin Cancer Res 2021;27:2899-909. [Crossref] [PubMed]
  81. Cho BC, Doebele RC, Lin J, et al. MA11.07 Phase 1/2 TRIDENT-1 Study of Repotrectinib in Patients with ROS1+ or NTRK+ Advanced Solid Tumors. J Thorac Oncol 2021;16:S174-5. [Crossref]
  82. Wolf J, Seto T, Han JY, et al. Capmatinib in MET Exon 14-Mutated or MET-Amplified Non-Small-Cell Lung Cancer. N Engl J Med 2020;383:944-57. [Crossref] [PubMed]
  83. Wolf J, Garon EB, Groen HJM, et al. Capmatinib in MET exon 14-mutated, advanced NSCLC: Updated results from the GEOMETRY mono-1 study. J Clin Oncol 2021;39:9020. [Crossref]
  84. Paik PK, Felip E, Veillon R, et al. Tepotinib in Non-Small-Cell Lung Cancer with MET Exon 14 Skipping Mutations. N Engl J Med 2020;383:931-43. [Crossref] [PubMed]
  85. Garon EB, Heist RS, Seto T, et al. Abstract CT082: Capmatinib in METex14-mutated (mut) advanced non-small cell lung cancer (NSCLC): Results from the phase II GEOMETRY mono-1 study, including efficacy in patients (pts) with brain metastases (BM). Cancer Res 2020;80:CT082. [Crossref]
  86. Lu S, Fang J, Li X, et al. Once-daily savolitinib in Chinese patients with pulmonary sarcomatoid carcinomas and other non-small-cell lung cancers harbouring MET exon 14 skipping alterations: a multicentre, single-arm, open-label, phase 2 study. Lancet Respir Med 2021;9:1154-64. [Crossref] [PubMed]
  87. Drilon A, Oxnard GR, Tan DSW, et al. Efficacy of Selpercatinib in RET Fusion-Positive Non-Small-Cell Lung Cancer. N Engl J Med 2020;383:813-24. [Crossref] [PubMed]
  88. Gainor JF, Curigliano G, Kim DW, et al. Pralsetinib for RET fusion-positive non-small-cell lung cancer (ARROW): a multi-cohort, open-label, phase 1/2 study. Lancet Oncol 2021;22:959-69. [Crossref] [PubMed]
  89. Drilon AE, Zhai D, Rogers E, et al. The next-generation RET inhibitor TPX-0046 is active in drug-resistant and naïve RET-driven cancer models. J Clin Oncol 2020;38:3616. [Crossref]
  90. Schoffski P, Cho BC, Italiano A, et al. BOS172738, a highly potent and selective RET inhibitor, for the treatment of RET-altered tumors including RET-fusion+ NSCLC and RET-mutant MTC: Phase 1 study results. J Clin Oncol 2021;39:3008. [Crossref]
  91. Planchard D, Smit EF, Groen HJM, et al. Dabrafenib plus trametinib in patients with previously untreated BRAF(V600E)-mutant metastatic non-small-cell lung cancer: an open-label, phase 2 trial. Lancet Oncol 2017;18:1307-16. [Crossref] [PubMed]
  92. Planchard D, Besse B, Groen HJM, et al. Dabrafenib plus trametinib in patients with previously treated BRAF(V600E)-mutant metastatic non-small cell lung cancer: an open-label, multicentre phase 2 trial. Lancet Oncol 2016;17:984-93. [Crossref] [PubMed]
  93. Planchard D, Kim TM, Mazieres J, et al. Dabrafenib in patients with BRAF(V600E)-positive advanced non-small-cell lung cancer: a single-arm, multicentre, open-label, phase 2 trial. Lancet Oncol 2016;17:642-50. [Crossref] [PubMed]
  94. Hong DS, DuBois SG, Kummar S, et al. Larotrectinib in patients with TRK fusion-positive solid tumours: a pooled analysis of three phase 1/2 clinical trials. Lancet Oncol 2020;21:531-40. [Crossref] [PubMed]
  95. Demetri GD, De Braud F, Drilon A, et al. Updated Integrated Analysis of the Efficacy and Safety of Entrectinib in Patients With NTRK Fusion-Positive Solid Tumors. Clin Cancer Res 2022;28:1302-12. [Crossref] [PubMed]
  96. Harada G, Choudhury NJ, Schram AM, et al. Mechanisms of acquired resistance to TRK inhibitors. J Clin Oncol 2022;40:3104. [Crossref]
  97. Katayama R, Gong B, Togashi N, et al. The new-generation selective ROS1/NTRK inhibitor DS-6051b overcomes crizotinib resistant ROS1-G2032R mutation in preclinical models. Nat Commun 2019;10:3604. [Crossref] [PubMed]
  98. Drilon A, Nagasubramanian R, Blake JF, et al. A Next-Generation TRK Kinase Inhibitor Overcomes Acquired Resistance to Prior TRK Kinase Inhibition in Patients with TRK Fusion-Positive Solid Tumors. Cancer Discov 2017;7:963-72. [Crossref] [PubMed]
  99. Tsuchida N, Murugan AK, Grieco M. Kirsten Ras* oncogene: significance of its discovery in human cancer research. Oncotarget 2016;7:46717-33. [Crossref] [PubMed]
  100. Ostrem JM, Peters U, Sos ML, et al. K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature 2013;503:548-51. [Crossref] [PubMed]
  101. Skoulidis F, Li BT, Dy GK, et al. Sotorasib for Lung Cancers with KRAS p.G12C Mutation. N Engl J Med 2021;384:2371-81. [Crossref] [PubMed]
  102. de Langen AJ, Johnson ML, Mazieres J, et al. Sotorasib versus docetaxel for previously treated non-small-cell lung cancer with KRASG12C mutation: a randomised, open-label, phase 3 trial. Lancet 2023;401:733-46. [Crossref] [PubMed]
  103. Jänne PA, Riely GJ, Gadgeel SM, et al. Adagrasib in Non-Small-Cell Lung Cancer Harboring a KRAS(G12C) Mutation. N Engl J Med 2022;387:120-31. [Crossref] [PubMed]
  104. Zhao Y, Murciano-Goroff YR, Xue JY, et al. Diverse alterations associated with resistance to KRAS(G12C) inhibition. Nature 2021;599:679-83. [Crossref] [PubMed]
  105. Awad MM, Liu S, Rybkin II, et al. Acquired Resistance to KRAS(G12C) Inhibition in Cancer. N Engl J Med 2021;384:2382-93. [Crossref] [PubMed]
  106. Wang X, Allen S, Blake JF, et al. Identification of MRTX1133, a Noncovalent, Potent, and Selective KRAS(G12D) Inhibitor. J Med Chem 2022;65:3123-33. [Crossref] [PubMed]
  107. Knox JE, Jiang J, Burnett GL, et al. Abstract 3596: RM-036, a first-in-class, orally-bioavailable, Tri-Complex covalent KRASG12D(ON) inhibitor, drives profound anti-tumor activity in KRASG12D mutant tumor models. Cancer Res 2022;82:3596. [Crossref]
  108. Mao Z, Xiao H, Shen P, et al. KRAS(G12D) can be targeted by potent inhibitors via formation of salt bridge. Cell Discov 2022;8:5. [Crossref] [PubMed]
  109. Koltun E, Cregg J, Rice MA, et al. Abstract 1260: First-in-class, orally bioavailable KRASG12V(ON) tri-complex inhibitors, as single agents and in combinations, drive profound anti-tumor activity in preclinical models of KRASG12V mutant cancers. Cancer Res 2021;81:1260. [Crossref]
  110. Hofmann MH, Lu H, Duenzinger U, et al. Abstract CT210: Trial in Process: Phase 1 studies of BI 1701963, a SOS1::KRAS Inhibitor, in combination with MEK inhibitors, irreversible KRASG12C inhibitors or irinotecan. Cancer Res 2021;81:CT210. [Crossref]
  111. Zhou C, Ramalingam SS, Kim TM, et al. Treatment Outcomes and Safety of Mobocertinib in Platinum-Pretreated Patients With EGFR Exon 20 Insertion–Positive Metastatic Non–Small Cell Lung Cancer: A Phase 1/2 Open-label Nonrandomized Clinical Trial. JAMA Oncol 2021;7:e214761. [Crossref] [PubMed]
  112. Park K, Haura EB, Leighl NB, et al. Amivantamab in EGFR Exon 20 Insertion-Mutated Non-Small-Cell Lung Cancer Progressing on Platinum Chemotherapy: Initial Results From the CHRYSALIS Phase I Study. J Clin Oncol 2021;39:3391-402. [Crossref] [PubMed]
  113. Mazieres J, Lafitte C, Ricordel C, et al. Combination of Trastuzumab, Pertuzumab, and Docetaxel in Patients With Advanced Non-Small-Cell Lung Cancer Harboring HER2 Mutations: Results From the IFCT-1703 R2D2 Trial. J Clin Oncol 2022;40:719-28. [Crossref] [PubMed]
  114. Le X, Cornelissen R, Garassino M, et al. Poziotinib in Non-Small-Cell Lung Cancer Harboring HER2 Exon 20 Insertion Mutations After Prior Therapies: ZENITH20-2 Trial. J Clin Oncol 2022;40:710-8. [Crossref] [PubMed]
  115. Zhou C, Li X, Wang Q, et al. Pyrotinib in HER2-Mutant Advanced Lung Adenocarcinoma After Platinum-Based Chemotherapy: A Multicenter, Open-Label, Single-Arm, Phase II Study. J Clin Oncol 2020;38:2753-61. [Crossref] [PubMed]
  116. Li BT, Shen R, Buonocore D, et al. Ado-Trastuzumab Emtansine for Patients With HER2-Mutant Lung Cancers: Results From a Phase II Basket Trial. J Clin Oncol 2018;36:2532-7. [Crossref] [PubMed]
  117. Ogitani Y, Aida T, Hagihara K, et al. DS-8201a, A Novel HER2-Targeting ADC with a Novel DNA Topoisomerase I Inhibitor, Demonstrates a Promising Antitumor Efficacy with Differentiation from T-DM1. Clin Cancer Res 2016;22:5097-108. [Crossref] [PubMed]
  118. Li BT, Smit EF, Goto Y, et al. Trastuzumab Deruxtecan in HER2-Mutant Non-Small-Cell Lung Cancer. N Engl J Med 2022;386:241-51. [Crossref] [PubMed]
  119. Schram AM, Odintsov I, Espinosa-Cotton M, et al. Zenocutuzumab, a HER2xHER3 Bispecific Antibody, Is Effective Therapy for Tumors Driven by NRG1 Gene Rearrangements. Cancer Discov 2022;12:1233-47. [Crossref] [PubMed]
  120. Schram AM, O'Reilly EM, O'Kane GM, et al. Efficacy and safety of zenocutuzumab in advanced pancreas cancer and other solid tumors harboring NRG1 fusions. J Clin Oncol 2021;39:3003. [Crossref]
  121. Carrizosa DR, Burkard ME, Elamin YY, et al. CRESTONE: Initial efficacy and safety of seribantumab in solid tumors harboring NRG1 fusions. J Clin Oncol 2022;40:3006. [Crossref]
  122. Heist RS, Guarino MJ, Masters G, et al. Therapy of Advanced Non-Small-Cell Lung Cancer With an SN-38-Anti-Trop-2 Drug Conjugate, Sacituzumab Govitecan. J Clin Oncol 2017;35:2790-7. [Crossref] [PubMed]
  123. Garon E, Johnson M, Lisberg A, et al. MA03.02 TROPION-PanTumor01: Updated Results From the NSCLC Cohort of the Phase 1 Study of Datopotamab Deruxtecan in Solid Tumors. J Thorac Oncol 2021;16:S892-3. [Crossref]
  124. Garon EB, Johnson ML, Lisberg AE, et al. LBA49 Efficacy of datopotamab deruxtecan (Dato-DXd) in patients (pts) with advanced/metastatic (adv/met) non-small cell lung cancer (NSCLC) and actionable genomic alterations (AGAs): Preliminary results from the phase I TROPION-PanTumor01 study. Ann Oncol 2021;32:S1326-7. [Crossref]
  125. Gazzah A, Ricordel C, Cousin S, et al. Efficacy and safety of the antibody-drug conjugate (ADC) SAR408701 in patients (pts) with non-squamous non-small cell lung cancer (NSQ NSCLC) expressing carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5). J Clin Oncol 2020;38:9505. [Crossref]
  126. Reck M, Rodríguez-Abreu D, Robinson AG, et al. Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. N Engl J Med 2016;375:1823-33. [Crossref] [PubMed]
  127. Herbst RS, Giaccone G, de Marinis F, et al. Atezolizumab for First-Line Treatment of PD-L1-Selected Patients with NSCLC. N Engl J Med 2020;383:1328-39. [Crossref] [PubMed]
  128. Sezer A, Kilickap S, Gümüş M, et al. Cemiplimab monotherapy for first-line treatment of advanced non-small-cell lung cancer with PD-L1 of at least 50%: a multicentre, open-label, global, phase 3, randomised, controlled trial. Lancet 2021;397:592-604. [Crossref] [PubMed]
  129. Reck M, Rodríguez-Abreu D, Robinson AG, et al. Five-Year Outcomes With Pembrolizumab Versus Chemotherapy for Metastatic Non-Small-Cell Lung Cancer With PD-L1 Tumor Proportion Score ≥ 50. J Clin Oncol 2021;39:2339-49. [Crossref] [PubMed]
  130. Mok TSK, Wu YL, Kudaba I, et al. Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): a randomised, open-label, controlled, phase 3 trial. Lancet 2019;393:1819-30. [Crossref] [PubMed]
  131. Gandhi L, Rodríguez-Abreu D, Gadgeel S, et al. Pembrolizumab plus Chemotherapy in Metastatic Non-Small-Cell Lung Cancer. N Engl J Med 2018;378:2078-92. [Crossref] [PubMed]
  132. Gadgeel S, Rodríguez-Abreu D, Speranza G, et al. Updated Analysis From KEYNOTE-189: Pembrolizumab or Placebo Plus Pemetrexed and Platinum for Previously Untreated Metastatic Nonsquamous Non-Small-Cell Lung Cancer. J Clin Oncol 2020;38:1505-17. [Crossref] [PubMed]
  133. Paz-Ares L, Luft A, Vicente D, et al. Pembrolizumab plus Chemotherapy for Squamous Non-Small-Cell Lung Cancer. N Engl J Med 2018;379:2040-51. [Crossref] [PubMed]
  134. Socinski MA, Jotte RM, Cappuzzo F, et al. Atezolizumab for First-Line Treatment of Metastatic Nonsquamous NSCLC. N Engl J Med 2018;378:2288-301. [Crossref] [PubMed]
  135. Nishio M, Barlesi F, West H, et al. Atezolizumab Plus Chemotherapy for First-Line Treatment of Nonsquamous NSCLC: Results From the Randomized Phase 3 IMpower132 Trial. J Thorac Oncol 2021;16:653-64. [Crossref] [PubMed]
  136. West H, McCleod M, Hussein M, et al. Atezolizumab in combination with carboplatin plus nab-paclitaxel chemotherapy compared with chemotherapy alone as first-line treatment for metastatic non-squamous non-small-cell lung cancer (IMpower130): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol 2019;20:924-37. [Crossref] [PubMed]
  137. Jotte R, Cappuzzo F, Vynnychenko I, et al. Atezolizumab in Combination With Carboplatin and Nab-Paclitaxel in Advanced Squamous NSCLC (IMpower131): Results From a Randomized Phase III Trial. J Thorac Oncol 2020;15:1351-60. [Crossref] [PubMed]
  138. Yang Y, Wang Z, Fang J, et al. Efficacy and Safety of Sintilimab Plus Pemetrexed and Platinum as First-Line Treatment for Locally Advanced or Metastatic Nonsquamous NSCLC: a Randomized, Double-Blind, Phase 3 Study (Oncology pRogram by InnovENT anti-PD-1-11). J Thorac Oncol 2020;15:1636-46. [Crossref] [PubMed]
  139. Zhou C, Wu L, Fan Y, et al. Sintilimab Plus Platinum and Gemcitabine as First-Line Treatment for Advanced or Metastatic Squamous NSCLC: Results From a Randomized, Double-Blind, Phase 3 Trial (ORIENT-12). J Thorac Oncol 2021;16:1501-11. [Crossref] [PubMed]
  140. Lu S, Wang J, Yu Y, et al. Tislelizumab Plus Chemotherapy as First-Line Treatment for Locally Advanced or Metastatic Nonsquamous NSCLC (RATIONALE 304): A Randomized Phase 3 Trial. J Thorac Oncol 2021;16:1512-22. [Crossref] [PubMed]
  141. Wang J, Lu S, Yu X, et al. Tislelizumab Plus Chemotherapy vs. Chemotherapy Alone as First-line Treatment for Advanced Squamous Non-Small-Cell Lung Cancer: A Phase 3 Randomized Clinical Trial. JAMA Oncol 2021;7:709-17. [Crossref] [PubMed]
  142. Zhou C, Chen G, Huang Y, et al. Camrelizumab plus carboplatin and pemetrexed versus chemotherapy alone in chemotherapy-naive patients with advanced non-squamous non-small-cell lung cancer (CameL): a randomised, open-label, multicentre, phase 3 trial. Lancet Respir Med 2021;9:305-14. [Crossref] [PubMed]
  143. Ren S, Chen J, Xu X, et al. Camrelizumab Plus Carboplatin and Paclitaxel as First-Line Treatment for Advanced Squamous NSCLC (CameL-Sq): A Phase 3 Trial. J Thorac Oncol 2022;17:544-57. [Crossref] [PubMed]
  144. Zhou C, Wang Z, Sun Y, et al. Sugemalimab versus placebo, in combination with platinum-based chemotherapy, as first-line treatment of metastatic non-small-cell lung cancer (GEMSTONE-302): interim and final analyses of a double-blind, randomised, phase 3 clinical trial. Lancet Oncol 2022;23:220-33. [Crossref] [PubMed]
  145. Hellmann MD, Paz-Ares L, Bernabe Caro R, et al. Nivolumab plus Ipilimumab in Advanced Non-Small-Cell Lung Cancer. N Engl J Med 2019;381:2020-31. [Crossref] [PubMed]
  146. Paz-Ares L, Ciuleanu TE, Cobo M, et al. First-line nivolumab plus ipilimumab combined with two cycles of chemotherapy in patients with non-small-cell lung cancer (CheckMate 9LA): an international, randomised, open-label, phase 3 trial. Lancet Oncol 2021;22:198-211. [Crossref] [PubMed]
  147. Rizvi NA, Cho BC, Reinmuth N, et al. Durvalumab With or Without Tremelimumab vs. Standard Chemotherapy in First-line Treatment of Metastatic Non-Small Cell Lung Cancer: The MYSTIC Phase 3 Randomized Clinical Trial. JAMA Oncol 2020;6:661-74. [Crossref] [PubMed]
  148. de Castro G Jr, Rizvi NA, Schmid P, et al. NEPTUNE: Phase 3 Study of First-Line Durvalumab Plus Tremelimumab in Patients With Metastatic NSCLC. J Thorac Oncol 2023;18:106-19. [Crossref] [PubMed]
  149. Mok T, Schmid P, de Castro G, et al. 482TiP Global, Phase 3 study of first-line durvalumab (MEDI4736) + tremelimumab vs. standard of care platinum-based chemotherapy in advanced/metastatic NSCLC: NEPTUNE. Ann Oncol 2016;27:ix155. [Crossref]
  150. Johnson M, Cho BC, Luft A, et al. PL02.01 Durvalumab ± Tremelimumab + chemotherapy as First-line Treatment for mNSCLC: Results from the Phase 3 POSEIDON Study. J Thorac Oncol 2021;16:S844. [Crossref]
  151. Boyer M, Şendur MAN, Rodríguez-Abreu D, et al. Pembrolizumab Plus Ipilimumab or Placebo for Metastatic Non-Small-Cell Lung Cancer With PD-L1 Tumor Proportion Score ≥ 50%: Randomized, Double-Blind Phase III KEYNOTE-598 Study. J Clin Oncol 2021;39:2327-38. [Crossref] [PubMed]
  152. Johnston RJ, Comps-Agrar L, Hackney J, et al. The immunoreceptor TIGIT regulates antitumor and antiviral CD8(+) T cell effector function. Cancer Cell 2014;26:923-37. [Crossref] [PubMed]
  153. Cho BC, Abreu DR, Hussein M, et al. Tiragolumab plus atezolizumab versus placebo plus atezolizumab as a first-line treatment for PD-L1-selected non-small-cell lung cancer (CITYSCAPE): primary and follow-up analyses of a randomised, double-blind, phase 2 study. Lancet Oncol 2022;23:781-92. [Crossref] [PubMed]
  154. Roche reports interim results for phase III SKYSCRAPER-01 study in PD-L1-high metastatic non-small cell lung cancer. Available online: https://www.roche.ch/com/media/releases/med-cor-2022-05-11.htm
  155. Ren S, Xiong X, You H, et al. The Combination of Immune Checkpoint Blockade and Angiogenesis Inhibitors in the Treatment of Advanced Non-Small Cell Lung Cancer. Front Immunol 2021;12:689132. [Crossref] [PubMed]
  156. Finn RS, Qin S, Ikeda M, et al. Atezolizumab plus Bevacizumab in Unresectable Hepatocellular Carcinoma. N Engl J Med 2020;382:1894-905. [Crossref] [PubMed]
  157. Rini BI, Plimack ER, Stus V, et al. Pembrolizumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N Engl J Med 2019;380:1116-27. [Crossref] [PubMed]
  158. Motzer R, Alekseev B, Rha SY, et al. Lenvatinib plus Pembrolizumab or Everolimus for Advanced Renal Cell Carcinoma. N Engl J Med 2021;384:1289-300. [Crossref] [PubMed]
  159. Yang JCH, Luft A, De La Mora Jiménez E, et al. 120O Pembrolizumab (Pembro) with or without lenvatinib (Lenva) in first-line metastatic NSCLC with PD-L1 TPS ≥1% (LEAP-007): A phase III, randomized, double-blind study. Ann Oncol 2021;32:S1429-30. [Crossref]
  160. Reckamp KL, Redman MW, Dragnev KH, et al. Phase II Randomized Study of Ramucirumab and Pembrolizumab Versus Standard of Care in Advanced Non-Small-Cell Lung Cancer Previously Treated With Immunotherapy-Lung-MAP S1800A. J Clin Oncol 2022;40:2295-306. [Crossref] [PubMed]
  161. Vansteenkiste JF, Cho BC, Vanakesa T, et al. Efficacy of the MAGE-A3 cancer immunotherapeutic as adjuvant therapy in patients with resected MAGE-A3-positive non-small-cell lung cancer (MAGRIT): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2016;17:822-35. [Crossref] [PubMed]
  162. Butts C, Socinski MA, Mitchell PL, et al. Tecemotide (L-BLP25) versus placebo after chemoradiotherapy for stage III non-small-cell lung cancer (START): a randomised, double-blind, phase 3 trial. Lancet Oncol 2014;15:59-68. [Crossref] [PubMed]
  163. Giaccone G, Felip E, Cobo M, et al. 1260MO Activity of OSE-2101 in HLA-A2+ non-small cell lung cancer (NSCLC) patients after failure to immune checkpoint inhibitors (ICI): Step 1 results of phase III ATALANTE-1 randomised trial. Ann Oncol 2020;31:S814-5. [Crossref]
  164. García-Pardo M, Gorria T, Malenica I, et al. Vaccine Therapy in Non-Small Cell Lung Cancer. Vaccines (Basel) 2022;10:740. [Crossref] [PubMed]
  165. Ott PA, Hu-Lieskovan S, Chmielowski B, et al. A Phase Ib Trial of Personalized Neoantigen Therapy Plus Anti-PD-1 in Patients with Advanced Melanoma, Non-small Cell Lung Cancer, or Bladder Cancer. Cell 2020;183:347-362.e24. [Crossref] [PubMed]
  166. Bear AS, Fraietta JA, Narayan VK, et al. Adoptive Cellular Therapy for Solid Tumors. Am Soc Clin Oncol Educ Book 2021;41:57-65. [Crossref] [PubMed]
  167. Creelan BC, Wang C, Teer JK, et al. Tumor-infiltrating lymphocyte treatment for anti-PD-1-resistant metastatic lung cancer: a phase 1 trial. Nat Med 2021;27:1410-8. [Crossref] [PubMed]
  168. Zhong S, Cui Y, Liu Q, et al. CAR-T cell therapy for lung cancer: a promising but challenging future. J Thorac Dis 2020;12:4516-21. [Crossref] [PubMed]
  169. Lu Y, Xue J, Deng T, et al. Safety and feasibility of CRISPR-edited T cells in patients with refractory non-small-cell lung cancer. Nat Med 2020;26:732-40. [Crossref] [PubMed]
  170. Grosser R, Cherkassky L, Chintala N, et al. Combination Immunotherapy with CAR T Cells and Checkpoint Blockade for the Treatment of Solid Tumors. Cancer Cell 2019;36:471-82. [Crossref] [PubMed]
  171. Adusumilli PS, Zauderer MG, Rivière I, et al. A Phase I Trial of Regional Mesothelin-Targeted CAR T-cell Therapy in Patients with Malignant Pleural Disease, in Combination with the Anti-PD-1 Agent Pembrolizumab. Cancer Discov 2021;11:2748-63. [Crossref] [PubMed]
  172. Davar D, Ding F, Saul M, et al. High-dose interleukin-2 (HD IL-2) for advanced melanoma: a single center experience from the University of Pittsburgh Cancer Institute. J Immunother Cancer 2017;5:74. [Crossref] [PubMed]
  173. Klapper JA, Downey SG, Smith FO, et al. High-dose interleukin-2 for the treatment of metastatic renal cell carcinoma: a retrospective analysis of response and survival in patients treated in the surgery branch at the National Cancer Institute between 1986 and 2006. Cancer 2008;113:293-301. [Crossref] [PubMed]
  174. Charych DH, Hoch U, Langowski JL, et al. NKTR-214, an Engineered Cytokine with Biased IL-2 Receptor Binding, Increased Tumor Exposure, and Marked Efficacy in Mouse Tumor Models. Clin Cancer Res 2016;22:680-90. [Crossref] [PubMed]
  175. Diab A, Tannir NM, Bentebibel SE, et al. Bempegaldesleukin (NKTR-214) plus Nivolumab in Patients with Advanced Solid Tumors: Phase I Dose-Escalation Study of Safety, Efficacy, and Immune Activation (PIVOT-02). Cancer Discov 2020;10:1158-73. [Crossref] [PubMed]
  176. Wrangle JM, Velcheti V, Patel MR, et al. ALT-803, an IL-15 superagonist, in combination with nivolumab in patients with metastatic non-small cell lung cancer: a non-randomised, open-label, phase 1b trial. Lancet Oncol 2018;19:694-704. [Crossref] [PubMed]
  177. Ridker PM, MacFadyen JG, Thuren T, et al. Effect of interleukin-1β inhibition with canakinumab on incident lung cancer in patients with atherosclerosis: exploratory results from a randomised, double-blind, placebo-controlled trial. Lancet 2017;390:1833-42. [Crossref] [PubMed]
  178. Paz-Ares L, Goto Y, Lim WDT, et al. 1194MO Canakinumab (CAN) + docetaxel (DTX) for the second- or third-line (2/3L) treatment of advanced non-small cell lung cancer (NSCLC): CANOPY-2 phase III results. Ann Oncol 2021;32:S953-S954. [Crossref]
  179. Available online: https://www.novartis.com/news/media-releases/novartis-top-line-results-canopy-1-phase-iii-study-support-further-evaluation-canakinumab-lung-cancer
  180. Hellmann M, Jänne P, Opyrchal M, et al. OA05.01 Efficacy/Safety of Entinostat (ENT) and Pembrolizumab (PEMBRO) in NSCLC Patients Previously Treated with Anti-PD-(L)1 Therapy. J Thorac Oncol 2018;13:S330. [Crossref]
  181. Gray JE, Saltos A, Tanvetyanon T, et al. Phase I/Ib Study of Pembrolizumab Plus Vorinostat in Advanced/Metastatic Non-Small Cell Lung Cancer. Clin Cancer Res 2019;25:6623-32. [Crossref] [PubMed]
  182. Routy B, Le Chatelier E, Derosa L, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018;359:91-7. [Crossref] [PubMed]
Cite this article as: Li MSC, Mok KKS, Mok TSK. Developments in targeted therapy & immunotherapy—how non-small cell lung cancer management will change in the next decade: a narrative review. Ann Transl Med 2023;11(10):358. doi: 10.21037/atm-22-4444

Download Citation