Skip to main content
Genes & Diseases logoLink to Genes & Diseases
. 2024 Jul 11;12(3):101374. doi: 10.1016/j.gendis.2024.101374

Differences between lung adenocarcinoma and lung squamous cell carcinoma: Driver genes, therapeutic targets, and clinical efficacy

Yue Shen a,b, Jie-Qi Chen a,b, Xiang-Ping Li a,b,
PMCID: PMC11904499  PMID: 40083325

Abstract

With the rapid advancements in second-generation gene sequencing technologies, a growing number of driver genes and associated therapeutic targets have been unveiled for lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC). While they are clinically classified as non-small cell lung cancer (NSCLC), they display distinct genomic features and substantial variations in clinical efficacy, underscoring the need for particular attention. Hence, this review provides a comprehensive overview of the latest advancements in driver genes, epigenetic targets, chemotherapy, targeted therapy, and immunotherapy for LUAD and LUSC. Additionally, it delves into the distinctions in signaling pathways and pivotal facets of clinical management specific to these two categories of lung cancer. Moreover, we furnish pertinent details regarding clinical trials pertaining to driver genes and epigenetics, thus establishing a theoretical foundation for the realization of precision treatments for LUAD and LUSC.

Keywords: Clinical efficacy, Gene mutations, Lung adenocarcinoma (LUAD), Lung squamous cell carcinoma (LUSC), Therapeutic targets

Introduction

Lung cancer is the leading cause of cancer-related deaths worldwide,1 broadly classified into non-small cell lung cancer (80%) (NSCLC) and small cell lung cancer (20%). NSCLC can be further categorized into various subtypes, including lung adenocarcinoma (LUAD), lung squamous cell carcinoma (LUSC), adenosquamous carcinoma, large cell carcinoma, sarcomatoid carcinoma, etc.2 In clinical practice, the two most common pathological types of NSCLC are LUAD (40%) and LUSC (30%).3 LUAD originates from the pulmonary epithelium in the lung parenchyma and exhibits distinctive papillary structures and glandular features, while LUSC originates from the basal cells of the lung's respiratory airways and is characterized by keratinized regions and intercellular bridges.2 In most prior research and expert guidelines, LUAD and LUSC were often treated with the same chemotherapy regimens. However, following the JMDB study, it was determined that the chemotherapy approach for LUAD differs from that of LUSC.4 As precision medicine advances, specific treatments for LUAD and LUSC are rapidly evolving. Numerous clinical studies have revealed significant differences, particularly in the context of targeted therapies, notably epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs). The distinct driver genes associated with LUAD and LUSC can result in varying prognosis outcomes during clinical targeted therapy. Therefore, a comprehensive understanding of the driver genes for these two types of lung cancer is crucial for clinical practitioners to choose appropriate targeted therapies. Furthermore, the development of specific drugs targeting the significantly expressed gene targets or signaling pathways for both types will bring about substantial breakthroughs in the precision treatment of LUAD and LUSC. Additionally, differences exist between LUAD and LUSC in their immune treatment microenvironment characteristics. This review delineates the treatment targets for LUAD and LUSC into two main categories: common driver gene targets and epigenetic targets. It provides a comprehensive overview of the similarities and differences in treatment targets and clinical approaches, highlighting the promising applications of specific targets in targeted therapy and precision medicine.

Genomic characteristics of LUAD and LUSC

In cancer, somatic copy number alterations are a common genomic abnormality. The differing frequencies of somatic copy number alterations in certain chromosomal regions in LUAD and LUSC5,6 lead to significant differences in their genetic mutations. The oncogenes KRAS (Kirsten rat sarcoma viral oncogene homologue), EGFR, BRAF (B-Raf proto-oncogene, serine/threonine kinase), and ALK (anaplastic lymphoma kinase) mutations frequently occur in LUAD, but are rarely found in LUSC (Fig. 1). Conversely, tumor suppressor genes such as TP53, CDKN2A (cyclin dependent kinase inhibitor 2A), PTEN (phosphatase and tensin homolog), and NOTCH1 (neurogenic locus notch homolog protein 1) exhibit higher mutation frequencies in LUSC. Mutations in these genes lead to frequent alterations in downstream signaling pathways such as PI3K (phosphoinositide 3-kinase)/AKT (protein kinase B)/mTOR (mechanistic target of rapamycin), RAS/RAF/MEK (mitogen-activated extracellular signal-regulated kinase), JAK (Janus kinase)/STAT (signal transducer and activator of transcription), thereby promoting the occurrence and development of both LUAD and LUSC. It is noteworthy that due to the frequent co-occurrence of tumor suppressor gene mutations with other genes, the genomic characteristics of LUAD and LUSC may undergo further alterations.

Figure 1.

Figure 1

Comparison of targetable driver gene mutation frequencies in lung adenocarcinoma and lung squamous cell carcinoma. Common oncogene mutations in lung adenocarcinoma include KRAS (25%), EGFR (14%), ALK (8%), BRAF (7%), MET (5%), ROS1 (5%), RET (4%), HER2 (3%), etc. Main oncogene mutations in lung squamous cell carcinoma include FGFR1 amplification (20%), PIK3CA (12%), DDR2 (3%), etc. Genetic mutation data were sourced from The Cancer Genome Atlas database. ALK, anaplastic lymphoma kinase; BRAF, B-Raf proto-oncogene, serine/threonine kinase; DDR2, discoidin domain receptor 2; EGFR, epidermal growth factor receptor; HER2, human epidermal growth factor receptor 2; KRAS, Kirsten rat sarcoma viral oncogene homologue; MET, mesenchymal–epithelial transition; PIK3CA, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha; ROS1, ROS proto-oncogene 1; RET, rearranged during transfection.

Proto-oncogene targets: Directly targetable

Common proto-oncogene targets in LUAD

EGFR

EGFR, located on chromosome 7q11.2 in humans,7 is a member of the HER (human epidermal growth factor receptor) family. Compared with European patients, Asian LUAD patients exhibit a higher prevalence of EGFR gene mutations, reaching around 50%.8 These mutations are not limited to changes in DNA sequences, such as exon 19 deletions, but also encompass EGFR gene amplifications and elevated EGFR protein expression. It is worth noting that in elderly patients, higher EGFR protein expression levels are observed, whereas there is no such correlation with EGFR gene amplification.9 Furthermore, this study revealed an association between EGFR amplification and a poorer prognosis in LUSC patients. Peng and colleagues confirmed that LUAD patients with EGFR amplification often have TP53 mutations, and EGFR amplification is a risk factor for disease progression in patients with brain metastases.10 Research indicates that the third-generation TKI drug, osimertinib, can not only reverse resistance to gefitinib and erlotinib in LUAD (T790M mutation) but can also serve as a neoadjuvant therapy, increasing the rate of surgical resection for patients.11 Exon 19 deletions and L858R point mutations are the two most common activating mutation types and are often used as biomarkers for TKI-targeted therapy.12 For patients with EGFR exon 19 deletions and L858R point mutations, osimertinib and gefitinib combined with pemetrexed have demonstrated the longest progression-free survival (PFS).13 The limited sample size and prevalent use of non-targeted drugs as first-line treatment in clinical practice for EGFR-mutant LUSC patients raise controversy regarding the efficacy of EGFR-TKIs14 (Table S1). Currently, there have been four generations of targeted drugs developed for EGFR mutations. Among these, the fourth generation, including drugs like TQB3804, U3-1402, and BLU-945, is still in the clinical trial phase.15 According to the results published by the American Association for Cancer Research in 2022 and 2023, BLU-945 is one of the fastest-progressing fourth-generation EGFR-TKI drugs, and the successful development of fourth-generation EGFR drugs holds promise in addressing resistance issues associated with third-generation EGFR-TKIs.

KRAS

KRAS belongs to the RAS oncogene family and is located in the 12q12.1 region.16 In contrast to EGFR mutations, KRAS mutations have a higher prevalence in LUAD patients in Europe, reaching approximately 30%, as opposed to Asian patients.17 KRAS mutations typically occur in exon 2, 3, or 4 regions.18 Among them, the KRAS G12C mutation is the most common, accounting for over 50% of KRAS mutations. Other KRAS mutations include G12V, G12D, and G13C. Due to the complex structure and lack of distinct catalytic activity of KRAS protein 19, targeting KRAS protein has proven to be challenging. Currently, only targeted drugs for the KRAS G12C mutation, such as sotorasib and adagrasib, have received approval for market use (Table 1).20,21 When KRAS-mutated LUAD is accompanied by LKB1 (liver kinase B1)/STK11 (serine/threonine kinase 11) loss, it can transition to LUSC, a transformation that further leads to resistance to KRAS inhibitors.22 This indicates that KRAS-mutated LUSC demonstrates a reduced sensitivity to KRAS inhibitors. In addition to directly inhibiting KRAS function, promoting the degradation of the KRAS protein can also achieve therapeutic goals, as seen with LC-2,23 YN14,24 and MS21.25 Research by Kostyrko's team at the University of California, USA, has discovered that knocking out UHRF1 can inhibit the growth of KRAS-driven mouse lung cancer tumors, suggesting that UHRF1 may become a potential therapeutic intervention target for KRAS-driven cancers.26 However, these drugs and targets are still in the early stages of clinical and preclinical research and require further validation for their safety and effectiveness.

Table 1.

Targeted therapies for driver genes in lung adenocarcinoma and lung squamous cell carcinoma.

Target Drug Phase Clinical trial Treatment Sample Outcome/status
Sponsor
mOS (months) mPFS (months) ORR (%)
EGFR Gefitinib Listed NCT00322452 Gefitinib vs. carboplatin/paclitaxel 1329 21.6 vs. 21.9 9.6 vs. 6.3 71.2% vs. 47.3% AstraZeneca
Erlotinib Listed NCT01342965 Erlotinib vs. gemcitabine/cisplatin 217 26.3 vs. 25.5 11.0 vs. 5.5 62.7% vs. 33.6% Hoffmann-La Roche
Icotinib Listed NCT01719536 Icotinib vs. pemetrexed/cisplatin 296 NA 11.2 vs. 7.9 64.8% vs. 33.8% Betta Pharmaceuticals Co., Ltd.
Afatinib Listed NCT01121393 Afatinib vs. gemcitabine/cisplatin 364 22.1 vs. 22.2 11.0 vs. 5.6 66.9% vs. 23.0% Boehringer Ingelheim
Dacomitinib Listed NCT01774721 Dacomitinib vs. gefitinib 452 34.1 vs. 27.0 14.7 vs. 9.2 74.9% vs. 71.6% Pfizer
Osimertinib Listed NCT02151981 Osimertinib vs. pemetrexed + carboplatin/cisplatin 419 26.8 vs. 22.5 10.1 vs. 4.4 70.6% vs. 31.4% AstraZeneca
Almonertinib Listed NCT02981108 Almonertinib 364 31.5 12.4 65.6% Jiangsu Hansoh Pharmaceutical Co., Ltd.
Alflutinib Listed NCT03452592 Alflutinib 220 NA 9.6 74.0% Allist Pharmaceuticals, Inc.
Amivantamab Listed NCT02609776 Amivantamab 780 22.8 8.3 40.0% Janssen Research & Development, LLC
Mobocertinib Listed NCT02716116 Mobocertinib 334 24 7.3 28.0% Takeda
KRAS Sotorasib Listed NCT03600883 Sotorasib 126 6.8 12.5 37.1% Amgen
Adagrasib Listed NCT03785249 Adagrasib 116 12.6 6.5 42.9% Mirati Therapeutics Inc.
ALK Crizotinib Listed NCT00932893 Crizotinib vs. chemotherapy 347 21.7 vs. 21.9 7.7 vs. 3.0 65% vs. 20% Pfizer
Alectinib Listed NCT02075840 Alectinib vs. crizotinib 303 NR 34.8 vs. 10.9 82.9% vs. 75.5% Hoffmann-La Roche
Ceritinib Listed NCT01828099 Ceritinib vs. chemotherapy 376 NR vs. 26.2 16.6 vs. 8.1 NA Novartis Pharmaceuticals
Brigatinib Listed NCT02737501 Brigatinib vs. crizotinib 275 NR 24 vs. 11 74% vs. 62% Ariad Pharmaceuticals
Ensartinib Listed NCT02767804 Ensartinib vs. crizotinib 290 NR 25.8 vs. 12.7 74% vs. 67% Xcovery Holding Company, LLC
Lorlatinib listed NCT03052608 Lorlatinib vs. crizotinib 296 NR NR vs. 9.3 76% vs. 58% Pfizer
ROS1 Crizotinib Listed NCT00585195 Crizotinib 53 51.4 19.2 72.0% Pfizer
Entrectinib Listed NCT02097810 Entrectinib 161 NR 15.7 67.1% Hoffmann-La Roche
RET Selpercatinib Listed NCT03157128 Selpercatinib (previously treated) vs. selpercatinib (treatment-naive) 356 NR 24.9 vs. 22 61% vs. 84% Loxo Oncology, Inc
Pralsetinib Listed NCT03037385 Pralsetinib (previously treated) vs. pralsetinib (treatment-naive) 233 NR 16.5 vs. 13 NA Hoffmann-La Roche
MET Capmatinib Listed NCT02414139 Capmatinib (previously treated) vs. capmatinib (treatment-naive) 373 NR 5.4 vs. 12.4 NR Novartis Pharmaceuticals
Tepotinib Listed NCT02864992 Tepotinib 337 17.1 8.5 46.0% EMD Serono Research & Development Institute, Inc.
Savolitinib Listed NCT02897479 Savolitinib 76 12.5 6.8 42.9% Hutchison Medipharma Limited
Glumetinib Listed NCT04270591 Glumetinib 79 17.3 8.5 65.8% Haihe Biopharma Co., Ltd.
BRAF Dabrafenib Listed NCT01336634 Dabrafenib vs. dabrafenib + trametinib (previously treated BRAF V600E-mutant metastatic NSCLC) vs. dabrafenib + trametinib (previously untreated BRAF V600E-mutant metastatic NSCLC) 177 12.7 vs. 18.2 vs. 17.3 5.4 vs. 10.2 vs. 10.8 NA Novartis Pharmaceuticals
HER2 Fam-trastuzumab deruxtecan-nxki Listed NCT03505710 Fam-trastuzumab deruxtecan-nxki 91 18.6 8.2 55.0% AstraZeneca
PIK3CA Taselisib Phase II NCT02785913 Taselisib 31 5.9 2.9 4.8% SWOG Cancer Research Network
TOS-358 Phase I NCT05683418 TOS-358 241 NA NA NA Totus Medicines
Copanlisib Phase I NCT03735628 Copanlisib + nivolumab 16 NA NA NA Bayer
ASN-003 Phase I NCT02961283 ASN-003 24 NA NA NA Asana BioSciences
Buparlisib Phase I NCT02128724 Buparlisib + radiotherapy treatment 21 NA NA NA University of Oxford
FGFR1 Nintedanib Listed NCT00805194 Docetaxel ± nintedanib 1314 10.1 vs. 9.1 3.4 vs. 2.7 NA Boehringer Ingelheim
Anlotinib Listed NCT02388919 Anlotinib vs. placebo 439 9.63 vs. 6.30 5.37 vs. 1.40 9.18% vs. 0.70% Chia Tai Tianqing Pharmaceutical Group Co., Ltd.
Fexagratinib Phase II/III NCT02965378 Fexagratinib 43 7.5 2.7 7.0% SWOG Cancer Research Network
Erdafitinib Phase II NCT03827850 Erdafitinib 22 NA NA NA Lung Cancer Group Cologne
Dovitinib Phase II NCT01861197 Dovitinib 27 NA NA NA Samsung Medical Center
AMG479 Phase Ib/II NCT00807612 AMG 479 + paclitaxel and carboplatin 49 NA NA NA NantCell, Inc.
BGJ398 Phase I NCT01004224 BGJ398 208 NA NA NA Novartis Pharmaceuticals
LY2874455 Phase I NCT01212107 LY2874455 94 NA NA 0.0% Eli Lilly and Company
Rogaratinib Phase I NCT01976741 Rogaratinib 37 NA 2.80 NA Bayer
GSK3052230 Phase I NCT01868022 GSK3052230 + paclitaxel + carboplatin/docetaxel/pemetrexed + cisplatin 65 NA NA NA GlaxoSmithKline
DDR2 ∗Dasatinib Phase II NCT01491633 Dasatinib 5 NA NA NA Dana-Farber Cancer Institute
STK11 ∗Talazoparib Phase II NCT04173507 Talazoparib + avelumab 42 7.6 2.7 2% SWOG Cancer Research Network
∗TNG260 Phase I/II NCT05887492 TNG260 vs. TNG260 + pembrolizumab 126 NA NA NA Tango Therapeutics, Inc.
∗Daratumumab Phase II NCT05807048 Daratumumab 14 NA NA NA NYU Langone Health
KEAP1 ∗Telaglenastat Phase II NCT04265534 Pembrolizumab and chemotherapy ± telaglenastat 40 NA NA NA Calithera Biosciences, Inc
∗Sapanisertib Phase II NCT02417701 Sapanisertib (NFEL2 squamous) vs. sapanisertib (KEAP1 squamous) vs. sapanisertib (KRAS/NFE2L2 or KEAP1 NSCLC) 34 NA 8.9 vs. 3.7 vs. 2.1 25% vs. 16.7% vs. 0% Calithera Biosciences, Inc
TP53 Eprenetapopt Phase Ib NCT04383938 Eprenetapopt + pembrolizumab 37 NA NA NA Aprea Therapeutics
∗Adavosertib Phase II NCT02087176 Docetaxel ± adavosertib 48 NA NA 9.4% AstraZeneca
∗Milademetan Phase II NCT05012397 Milademetan 65 NA NA NA Rain Oncology Inc
PTEN ∗AZD8186 Phase I NCT01884285 AZD8186 ± abiraterone acetate/AZD2014 147 NA NA NA AstraZeneca
NOTCH1 ∗RO4929097 Phase II NCT01070927 RO4929097 7 NA NA NA Hoffmann-La Roche
CB-103 Phase I/II NCT03422679 CB-103 79 NA NA NA Cellestia Biotech AG
Crenigacestat Phase I NCT02836600 Crenigacestat 12 NA NA NA Eli Lilly and Company
Brontictuzumab Phase I NCT01778439 Brontictuzumab 48 NA NA NA OncoMed Pharmaceuticals, Inc.
CDKN2A ∗Palbociclib Phase II/III NCT02785939 Palbociclib 32 7.1 1.7 6.0% SWOG Cancer Research Network
∗ABT-348 Phase II NCT02478320 ABT-348 12 NA NA NA M.D. Anderson Cancer Center
∗Abemaciclib Phase II EUCTR2014-004832-20-DE Abemaciclib vs. docetaxel 150 NA NA NA Eli Lilly & Co.
∗PF-07248144 Phase I NCT04606446 PF-07248144 ± fulvestrant/letrozole + palbociclib/PF-07220060 + fulvestrant 186 NA NA NA Pfizer

Notes: The drug information and data in the table were sourced from Pharm Snap, ClinicalTrials.gov, and the EU Clinical Trials Register. Non-direct drugs are indicated by an asterisk (∗). Dasatinib, Bcr-Abl kinase inhibitor, Talazoparib, PARP1/PARP2 inhibitor; TNG260, HDAC1 inhibitor; Daratumumab, CD38 inhibitor; Telaglenastat, glutaminase 1 inhibitor; Sapanisertib, mTOR1/2 inhibitor; Adavosertib, Wee1 inhibitor; Milademetan, MDM2 inhibitor; AZD8186, PI3K inhibitor; RO4929097, γ-secretase inhibitor; Palbociclib, CDK4/6 inhibitor; ABT-348, aurora inhibitor; Abemaciclib, CDK4/6 inhibitor; PF-07248144, KAT6A/B inhibitor; NA, not available; NR, not reached; ORR, objective response rate; mOS, metastatic overall survival; mPFS, metastatic progression-free survival; EGFR, epidermal growth factor receptor; KRAS, Kirsten rat sarcoma viral oncogene homologue; MET, mesenchymal–epithelial transition; RET, rearranged during transfection; HER2, human epidermal growth factor receptor 2; ALK, anaplastic lymphoma kinase; ROS1, ROS proto-oncogene 1; BRAF, B-Raf proto-oncogene, serine/threonine kinase; PIK3CA, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha; DDR2, discoidin domain receptor 2; FGFR1, fibroblast growth factor receptor 1; STK11, serine/threonine kinase 11; KEAP1, Kelch-like ECH-associated protein 1; PTEN, phosphatase and tensin homolog; NOTCH1, neurogenic locus notch homolog protein 1; CDKN2A, cyclin dependent kinase inhibitor 2A; NSCLC, non-small cell lung cancer.

ALK

ALK originates from the insulin receptor superfamily.27 The most common ALK gene mutation is the EML4 (echinoderm microtubule associated protein like 4)-ALK fusion gene, accounting for over 85% of ALK mutations.28 In young, non-smoking patients with LUAD who do not have EGFR mutations, the prevalence of EML4-ALK fusion can reach 25%–30%.29 ALK-positive patients tend to have better treatment responses and survival rates compared with ALK-negative NSCLC patients, particularly evident in targeted therapies for ALK mutations. Research involving ALK-positive non-LUSC patients has shown that those treated with crizotinib achieved a significantly improved PFS of 10.9 months compared with conventional chemotherapy.30 Currently, the U.S. Food and Drug Administration (FDA) has approved five targeted drugs for ALK mutations, namely crizotinib, ceritinib, alectinib, brigatinib, and lorlatinib. According to the latest research results from the CROWN trial, researchers estimate that the metastatic PFS with the newly approved third-generation ALK inhibitor lorlatinib for treating ALK-mutated NSCLC will exceed 60 months.31 Lorlatinib remains effective for patients resistant to crizotinib and alectinib,32 with a high efficacy rate of up to 82% for patients with brain metastases.33

ROS1

Similar to the ALK gene, ROS1 (ROS proto-oncogene 1, receptor tyrosine kinase) also belongs to the insulin receptor family.34 ROS1 fusions can be categorized into CD74 (cluster of differentiation 74)-ROS1 (44%), EZR (Ezrin)-ROS1 (16%), SDC4 (syndecan 4)-ROS1 (14%), and SLC34A2 (solute carrier family 34 member 2)-ROS1 (10%).35 Due to the 70% amino acid sequence homology in the kinase domain between ROS1 and ALK, patients with ROS1 gene fusions tend to respond well to most ALK inhibitors.35,36 Therefore, patients with ROS1 rearrangements generally have a favorable prognosis similar to ALK-mutated patients. However, it is worth noting that LUAD patients carrying ROS1 fusion mutations have a higher risk of venous thrombosis.37,38 Currently, both crizotinib and entrectinib have been approved by the FDA for treating advanced NSCLC patients with ROS1 rearrangements. Unlike crizotinib, entrectinib can penetrate the blood–brain barrier, making it more effective for patients with brain metastases.39 However, selective TKIs specifically targeting ROS1 have not yet been developed.

RET

RET (rearranged during transfection) is one of the members of the calmodulin superfamily, and its mutations primarily occur in untreated, non-smoking young patients.40 Patients with RET mutations have a higher risk of brain metastasis and pleural dissemination.41,42 Interestingly, tumors with RET mutations tend to exhibit a higher sensitivity to regimens that include pemetrexed chemotherapy, possibly due to lower expression levels of thymidylate synthase in RET-mutated tumor tissues.43,44 Furthermore, in some studies, RET rearrangements have been identified as potential mechanisms of resistance to EGFR-TKIs in EGFR-mutated NSCLC.45,46 Currently, there are selective RET inhibitors available for the treatment of LUAD patients with RET rearrangements, such as selpercatinib and pralsetinib. Research has shown that selpercatinib and pralsetinib are highly effective in treating RET-positive LUAD patients with relatively few side effects.47,48

MET

MET (mesenchymal–epithelial transition) is located on chromosome 7q31 region.49 MET exon 14 skipping mutations exhibit some clinical heterogeneity and are commonly found in older, smoking history-associated LUAD patients.50,51 While the occurrence rate of MET mutations in LUAD is less than 5%,52 it can be as high as around 20% in patients who have shown resistance to molecular targeted therapies such as EGFR, ALK, and ROS1.53 MET amplification or excessive activation of the MET protein can lead to resistance to first-generation and third-generation EGFR-TKI drugs. However, the use of MET inhibitors has been shown to effectively overcome resistance to third-generation EGFR-TKIs.54 For example, in the ORCHARD trial, LUAD patients with EGFR and MET co-alterations who had become resistant to first-line TKI treatment achieved a disease control rate of 82% with the combination of savolitinib and osimertinib.55 Currently, drugs targeting MET mutations include crizotinib, savolitinib, capmatinib, and tepotinib. Capmatinib has shown activity against brain metastases as well.56

BRAF

BRAF, a specific protein kinase composed of 766 amino acids that phosphorylates threonine and serine residues,57 is located in the 7q34 region of chromosome 7.58 BRAF V600 mutations account for approximately 30%–50% of all BRAF mutations,59 and are predominantly found in non-smoking females.60 In contrast, non-V600 mutations are mainly observed in male smokers.61 BRAF mutations lead to reduced response to first-line platinum-based chemotherapy and are associated with poorer prognosis in LUAD patients.62 However, studies have indicated that BRAF mutations are associated with sensitivity to immune checkpoint inhibitor therapy.63, 64, 65 Because sustained activation of BRAF disrupts the MEK/ERK (extracellular signal-regulated kinase) signaling pathway, leading to excessive cell proliferation and malignancy66 (Fig. 2), clinical management of LUAD patients with BRAF mutations often involves combination therapy with BRAF and MEK inhibitors. Dual-target treatment with dabrafenib and trametinib has been well-validated for safety and efficacy in various clinical trials67,68 and is currently the only targeted therapy option for BRAF V600E-mutated NSCLC in China.

Figure 2.

Figure 2

Mechanisms of action of driver genes in lung adenocarcinoma. EGFR, HER2, ALK, ROS1, RET, and MET all belong to the RTK family. Aberrantly activated RTKs bind to Grb2, which recruits SOS proteins to form a complex, catalyzing the binding of KRAS to GTP. Activated KRAS protein can activate downstream signaling pathways including RAF/MEK/ERK, PI3K/AKT/mTOR, JAK/STAT, and Ral-GEF/TBK1/NF-κB, thereby promoting tumor cell survival and proliferation. In addition, ROS1 can activate the VAV3-RHO pathway, further enhancing tumor cell migration and invasion. ALK, anaplastic lymphoma kinase; AKT, protein kinase B; EGFR, epidermal growth factor receptor; EML4, echinoderm microtubule associated protein like 4; ERK, extracellular signal-regulated kinase; GDP, guanosine diphosphate; Grb2, growth factor receptor-bound protein 2; GTP, guanosine triphosphate; HER1/2/3/4, human epidermal growth factor receptor 1/2/3/4; JAK, Janus kinase; KRAS, Kirsten rat sarcoma viral oncogene homologue; MEK, mitogen-activated extracellular signal-regulated kinase; MET, mesenchymal–epithelial transition; mTOR, mechanistic target of rapamycin; NF-κB, nuclear factor-kappa B; PI3K, phosphoinositide 3-kinase; Ral-GEF, Ral guanine nucleotide exchange factors; RET, rearranged during transfection; RHO, rhodopsin, a family member of the small G protein superfamily; ROS1, ROS proto-oncogene 1; RTK, receptor tyrosine kinase; SOS, son of sevenless; STAT, signal transducer and activator of transcription; TBK1, TANK binding kinase 1; VAV3, Vav guanine nucleotide exchange factor 3, a guanine exchange factor for RHO.

HER2

HER2 (human epidermal growth factor receptor 2) mutations in LUAD primarily involve exon 20 insertion mutations.69 This mutation is more common in non-smoking females and Asians70 and is associated with a higher likelihood of brain metastasis occurrence compared with other gene mutation statuses.71 In the DESTINY-Lung01 trial, trastuzumab deruxtecan demonstrated durable anti-tumor activity (with a median survival of 17.8 months) and a manageable safety profile.72 Based on these results, the NCCN guidelines for NSCLC recommend the use of trastuzumab deruxtecan in the treatment of HER2-mutated lung cancer.73 Furthermore, some HER2-targeted drugs like lapatinib, neratinib, and tucatinib have shown promising efficacy in clinical settings. An EGFR/HER2 dual-targeted inhibitor called pyrotinib has exhibited stronger tumor-suppressive effects in advanced LUAD patients carrying HER2 exon 20 mutations,74 closely related to its comprehensive inhibition of the HER2 signaling pathway.

Common proto-oncogene targets in LUSC

PIK3CA

PIK3CA (phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha), located on chromosome 3, is a representative gene frequently implicated in LUSC, with abnormalities reported in approximately 35% of patients.75 Upon occurrence of typical mutations or amplifications in PIK3CA, aberrant activation of the PI3K/ATK/mTOR signaling pathway promotes cancer cell proliferation and invasion (Fig. 3). Moreover, PIK3CA mutations are associated with chemotherapy resistance and poor prognosis.76 In tackling PIK3CA mutations, interventions can target not only PIK3CA itself but also downstream components of the PI3K signaling pathway, such as AKT and mTOR. Currently, the targeted drug alpelisib for PIK3CA has received approval for breast cancer and has been the subject of numerous clinical trials in the field of LUSC. These trials include PIK3CA inhibitors alpelisib (BYL719), dactolisib (BEZ235), and GDC-0941, mTOR inhibitors everolimus (RAD001) and temsirolimus (CCI-779), and AKT inhibitors miransertib (ARQ-092) and ipatasertib (GDC-0068).

Figure 3.

Figure 3

Mechanisms of action of driver genes in lung squamous cell carcinoma. Upon abnormal activation of the FGFR1 receptor, various signaling pathways, including PLCγ/PKC, RAS/RAF/ERK, PI3K/AKT/mTOR, and JAK/STAT, are activated to promote tumor cell growth. Overexpression or mutations in DDR2 can activate the above-mentioned signaling pathways and activate the Lyn/SHIP2/ITSN pathway. The CDK2-CyclinE and CDK4/6-CyclinD complexes, regulated by the P53 target gene P21, jointly modulate the phosphorylation level of the Rb protein. Hypophosphorylated RB prevents cells from passing through the G1/S checkpoint (also known as the R point), causing cell arrest in the G1 phase. Up-regulation of CyclinD and mutant P53 can lead to the accumulation of unrepaired damaged DNA, ultimately resulting in tumorigenesis. NOTCH1 protein can be activated by binding to NOTCH ligands (Delta-like1/3/4 and Jagged1/2). The Notch receptor is initially cleaved by ADAM10 or TACE and then cleaved by γ-secretase, releasing NICD into the nucleus, where it binds to the transcription factor CSL and recruits MAML protein to form a complex that activates target genes such as HES and HEY. Among them, HES1 can inhibit the PTEN gene. The product encoded by the PTEN gene catalyzes the hydrolysis of PIP3 to PIP2, and its inactivation or mutation leads to activation of the PI3K/AKT pathway. NRF2, under steady-state conditions, binds to the protein KEAP1 and is degraded via the proteasome. When exposed to oxidative stress, KEAP1 undergoes a conformational change, releasing NRF2. NRF2 accumulates and translocates to the cell nucleus, where it binds to sMAF protein and activates the transcription of many genes encoding cellular protective factors. Mutations in KEAP1 lead to the activation of the Nrf2 pathway, promoting the survival of tumor cells. ADAM10, ADAM metallopeptidase domain 10; AKT, protein kinase B; CDK2/4/6, cyclin dependent kinase 2/4/6; CSL, CBF1, suppressor of hairless, lag-1; DDR2, discoidin domain receptor 2; ERK, extracellular signal-regulated kinase; FGFR1, fibroblast growth factor receptor 1; HES1, hairy and enhancer of split 1; ITSN, intersectin; JAK, Janus kinase; KEAP1, Kelch-like ECH-associated protein 1; Lyn, Lck/yes-related protein tyrosine kinase, a member of the Src protein tyrosine kinase family; MAML, mastermind-like; mTOR, mechanistic target of rapamycin; NICD, Notch intracellular domain; NOTCH1, neurogenic locus notch homolog protein 1; NRF2, nuclear factor erythroid 2-related factor 2; PKC, protein kinase C; PI3K, phosphoinositide 3-kinase; PIP2, phosphatidylinositol 4,5-bisphosphate; PIP3, phosphatidylinositol (3,4,5)-trisphosphate; PLCγ, phospholipase C gamma; PTEN, phosphatase and tensin homolog; SHIP2, SH2 domain-containing inositol polyphosphate 5′-phosphatase 2; sMaf, small musculoaponeurotic fibrosarcoma; STAT, signal transducer and activator of transcription; TACE, TNF-α converting enzyme.

FGFR1 amplification

FGFR1 amplification is the most common mutation type of FGFR (fibroblast growth factor receptor) in LUSC, accounting for approximately 40%, with an incidence rate of around 20%.77 FGFR1 amplification triggers the PLCγ (phospholipase C gamma)/PKC (protein kinase C), RAS/MAPK (mitogen-activated protein kinase), and PI3K/AKT pathways, promoting angiogenesis as well as the growth and proliferation of tumor cells.78 The influence of FGFR1 amplification on the prognosis for LUSC patients is subject to debate. While one study indicated that FGFR1 amplification was linked to poorer outcomes,79 another found it did not affect PFS and overall survival (OS) in LUSC patients but was strongly associated with lymph node metastasis.80 Consequently, whether FGFR1 amplification can serve as an independent prognostic marker for LUSC requires further research. Currently, various inhibitors targeting FGFR receptors have been developed and are primarily divided into two categories, small molecule TKIs such as AZD4547, BGJ398, and LY2874455, and FGFR antibodies such as FP1039, AMG479, and BIIB022. Unlike the promising results in preclinical models, many FGFR drugs have shown lower disease control rates in clinical trials for lung squamous cell carcinoma, such as AZD4547's phase Ib trial (NCT00979134),81 phase II trial (NCT02965378),82 rogaratinib's phase II study (NCT01976741),83 and BGJ398's phase I trial (NCT01004224).84 Therefore, considering the combination of FGFR1 inhibitors with chemotherapy or immunotherapy may hold promise in achieving breakthroughs in anti-tumor activity.

DDR2

DDR2 (discoidin domain receptor 2) is a receptor tyrosine kinase capable of binding to collagen.85 Research has shown that phosphorylated DDR2 in cancer cells can activate the JAK2 (Janus kinase 2)/ERK pathway and may initiate the Ras and PI3K pathways.86 In LUSC, about 4% of patients have DDR2 mutations. These mutations may lead to aberrations in downstream signaling pathways or affect the growth, migration, and invasion of LUSC through the promotion of epithelial–mesenchymal transition.87 Studies have indicated that BCR (breakpoint cluster region)-ABL kinase inhibitors used to treat chronic myeloid leukemia, such as dasatinib, nilotinib, and imatinib, have significant inhibitory effects on DDR2 kinase.88 In 2011, a phase II clinical study (NCT01491633) investigating the efficacy of dasatinib in advanced squamous cell carcinoma was conducted.89 However, because of patients' intolerance to the drug and associated toxicity issues, the trial was terminated prematurely in 2013. Currently, a potential drug targeting DDR2 mutations in squamous cell carcinoma (4-amino-6-(2,6-dichlorophenyl)-8-methyl-2-(phenylamino)-pyrido [2,3-d] pyrimidin-7(8H)) is in the preclinical trial stage.

Tumor suppressor gene targets: Difficult to directly target

Common tumor suppressor gene targets in LUAD

STK11/LKB1

STK11 encodes LKB1 and exhibits a specific high-frequency mutation in LUAD,90 often accompanied by KRAS mutation.91 Inactivation of STK11 up-regulates the expression of genes associated with angiogenesis and cell migration, thereby enhancing the invasiveness of LUAD.92 Despite this, standard treatments such as chemotherapy and immunotherapy tend to be ineffective in patients harboring STK11 mutations, compounding the challenge in treating such cases. Inactivation of STK11 can lead to aberrant activation of the downstream mTOR signaling pathway and affect glutamine metabolism.93 Therefore, mTOR and glutamine inhibitors (such as temsirolimus and everolimus) may have therapeutic effects on it. Furthermore, Shaw et al. found in animal experiments that the combination of the HDAC3 inhibitor entinostat and the MEK inhibitor trametinib effectively inhibits the progression of such tumors.94 At present, some non-directly targeted drugs for STK11 mutations have entered clinical trials (Table 1), but direct targeted drugs for STK11 are still in the research stage. Therefore, improving effective treatment for patients with this mutation remains an urgent clinical challenge.

KEAP1

Mutations in KEAP1 (Kelch-like ECH-associated protein 1) lead to activation of the Nrf2 (nuclear factor erythroid 2-related factor 2) pathway, thereby promoting tumor cell proliferation and survival (Fig. 2). KEAP1 mutations are commonly found in LUAD, while Nrf2 mutations are mainly observed in LUSC. Patients with KEAP1/NRF2 mutations in LUAD exhibit varying degrees of resistance to chemotherapy, radiotherapy, targeted therapy, and immunotherapy.95 Concurrent mutations in STK11 and KEAP1 up-regulate the iron death protective gene SCD (stearoyl-CoA desaturase), conferring resistance to drug-induced ferroptosis.96 KEAP1 deficiency also promotes the progression of KRAS-driven lung cancer and leads to its dependence on glutamine metabolism.97 Research has found that KEAP1 inactivation results in glucose dependence, rendering lung cancer cells sensitive to glucose inhibitors.98 Currently, therapeutic strategies targeting the KEAP/NRF2 signaling pathway primarily focus on glutaminase inhibition, glutamine antagonism, and PI3K-AKT signaling suppression. In preclinical studies, KEAP1 mutations have shown high selectivity and sensitivity to mTOR inhibitors (such as rapamycin and NVP-BEZ235).99 Additionally, clinical trials are underway for the combination therapy of the mTOR inhibitor TAK-228 with the glutaminase inhibitor CB-839 in the treatment of NSCLC.

Common tumor suppressor gene targets in LUSC

TP53

TP53 mutations are the most prevalent genetic aberrations in both LUAD and LUSC, and their occurrence is closely associated with smoking.100,101 TP53 mutations lead to impaired cellular apoptosis, DNA damage repair, and cell cycle regulation. Song et al. identified disparities in the TP53 mutation sites between LUAD and LUSC (Table S1). Furthermore, the detection of TP53 gene mutations in early-stage LUAD patients often portends a poorer OS, a conclusion not applicable to LUSC.102 Some studies suggest that the occurrence of TP53 mutations in LUSC is related to resistance to platinum-based chemotherapy drugs.103,104 This may be due to alterations in DNA repair mechanisms required for treatment. Besides, immune features associated with TP53 mutations, such as T cell infiltration and tumor mutation burden, may enhance responsiveness to immunotherapy.105 Presently, research on targeted therapies for TP53 mutations is still in its early stages. For example, APR-246 can restore normal function to mutated TP53 genes,106 while COTI-2 selectively kills tumor cells with TP53 mutations.107 Tumors with P53 mutations exhibit a heightened dependence on the G2/M checkpoint to maintain genomic stability, rendering them more susceptible to the effects of Wee1 inhibitors such as adavosertib.108 Nonetheless, TP53 mutation is not the most reliable predictive biomarker. This observation may account for the modest benefits observed in many studies when Wee1 inhibitors are combined with chemotherapy. Furthermore, inhibitors targeting TP53-protective proteins like Mdm2 and Mdm4 hold promise in restoring TP53 protein function.109,110 Similarly, cell cycle regulatory proteins such as CDK4/6 (cyclin dependent kinase 4/6), which are associated with TP53, may serve as potential therapeutic targets.111

PTEN

PTEN is a major negative regulator of the PI3K/AKT/mTOR signaling pathway. In LUSC, the mutation rate of PTEN is approximately 10%, with most mutations occurring in smokers.112 This may be related to smoking-induced down-regulation of PTEN expression through immune-mediated mechanisms. Studies have shown that low expression levels of PTEN protein are associated with decreased survival rates in lung cancer patients.113 This also underscores the close relationship between down-regulated PTEN protein levels and increased cancer susceptibility. Spoerke et al. discovered that NSCLC models with PTEN mutations exhibit high sensitivity to PI3K inhibitors.114 Similarly, PTEN inactivation can lead to resistance to PI3K inhibitors.115 Furthermore, the imbalance in the expression of PI3K and PTEN can promote immune evasion by tumors.116 However, currently, all PI3K/AKT inhibitors or mTOR inhibitors cannot distinguish between LUSC with PTEN loss and those without.

NOTCH1

NOTCH1 is a transmembrane receptor that plays a crucial regulatory role in controlling cell fate, cell proliferation, and differentiation.117 Research has revealed that NOTCH1 has a tumor-suppressive role in LUSC models but promotes tumor growth in LUAD.118 According to The Cancer Genome Atlas database, approximately 13% of LUSC patients have NOTCH1 gene mutations. Furthermore, studies by Yoshida and colleagues found that NOTCH1 mutations in lung cells are more common among individuals with a history of smoking compared with non-smokers.119 Recent research suggests that NOTCH1 gene mutations may predict the benefit of immunotherapy for LUSC patients.117,120 This may be linked to the activation of DNA damage response pathways and the immune microenvironment associated with NOTCH1 gene mutations. Since NOTCH1 can inhibit the P53 protein 121, blocking the transmission of the NOTCH1 signaling pathway can increase P53 stability, promoting cell apoptosis. Additionally, interfering with the abnormal activation of NOTCH signaling by inhibiting the key enzyme gamma-secretase is another approach. Some related gamma-secretase inhibitors include PF-03084014, RO4929097, and MK-0752. Furthermore, researchers are exploring antibodies targeting NOTCH1 in both research and clinical trials, such as tarextumab and OMP-52M51.

CDKN2A

CDKN2A is a tumor suppressor gene located on the short arm of chromosome 9. It encodes the proteins p16INK4a (P16) and p14ARF (P14).122,123 P16 binds to CDK4/6 and cyclin D, inhibiting the transition of the cell cycle from G1 to S phase, thereby affecting cell proliferation.124 Research has shown that high expression of P16 in LUSC patients is associated with favorable survival outcomes, although this trend is not observed in LUAD.125 CDK inhibitors such as flavopiridol and dinaciclib have demonstrated potential therapeutic effects by inducing cytotoxicity in CDKN2A-defective LUSC cells through enhanced apoptosis.126 Furthermore, there is a clinical case report of improved efficacy with the CDK4/6 inhibitor abemaciclib in a patient carrying mutations in CDKN2A, PI3K, and TP53 simultaneously.127 Currently, CDK4/6 inhibitors are primarily used in breast cancer treatment, and research on their use in LUSC is still in the early phases of phase I and phase II clinical trials.

Emerging driver gene targets: Epigenetic therapeutic targets

The term “epigenetics” was originally coined by Conrad Waddington, encompassing various aspects such as DNA methylation, histone modifications, non-coding RNAs, and chromatin 128. In tumor cells, epigenetic features are extensively dysregulated, driving the development of targeted epigenetic anti-cancer therapies. Regulators associated with DNA methylation and post-translational histone modifications hold promise as emerging driver gene targets in both LUAD and LUSC.129 Further studies have shown that downstream effects caused by genetic mutations may lead to different epigenetic modifications in LUAD and LUSC.92

Epigenetic therapy targets for LUAD

EZH2

EZH2 (enhancer of zeste homolog 2), a histone methyltransferase, serves as a critical subunit catalyzing the activity of PRC2 (polycomb repressive complex 2).130 Studies have indicated a correlation between elevated EZH2 expression and proliferation and invasion of LUAD cells,131 potentially due to its role in regulating the vascular endothelial growth factor-A (VEGF-A) signaling pathway and AKT phosphorylation mechanisms.132,133 Research by Fan and Kim et al. has shown a link between high EZH2 expression in LUAD, smoking, and poor prognosis, whereas there is no significant correlation in LUSC.134,135 It is worth noting that inhibiting EZH2 may lead to resistance to EGFR-TKIs in NSCLC,136 and impact the tumor microenvironment, enhancing anti-tumor immunity.130 Currently, several EZH2 inhibitors are undergoing clinical and preclinical studies, primarily for lymphoma treatment.137 For advanced solid tumors like SHR2554, it is in phase II clinical trials, and the effectiveness of combining EZH2 inhibitors with ICB is under clinical evaluation (Table 2).

Table 2.

Epigenetic targeted therapies for lung adenocarcinoma and lung squamous cell carcinoma.

Target Drug Indication Phase Clinical trial Treatment Sample Status Sponsor
EZH2 SHR-2554 Advanced solid tumor Phase Ⅰ/Ⅱ NCT04407741 SHR1701 ± SHR2554 100 Recruiting Chinese PLA General Hospital
Tulmimetostat Advanced solid tumor Phase Ⅰ/Ⅱ NCT04104776 Tulmimetostat 213 Recruiting Constellation Pharmaceuticals
Tazemetostat Advanced non-small cell lung cancer Phase Ⅰ/Ⅱ NCT05467748 Pembrolizumab + tazemetostat 66 Not yet recruiting VA Office of Research and Development
XNW-5004 Advanced solid tumor Phase I ChiCTR2100048401 XNW-5004 82 Recruiting Hematology Hospital of Chinese Academy of Medical Sciences
TR115 Advanced solid tumor Phase I NCT05650580 TR115 26 Recruiting Tarapeutics Science Inc.
BRD4 RNK05047 Advanced solid tumors Phase Ⅰ/Ⅱ NCT05487170 RNK05047 105 Recruiting Ranok Therapuetics Co. Ltd.
NHWD-870 NSCLC Phase I CTR20202650 NHWD-870 30 Not yet recruiting Hunan Hengya Pharmaceutical Technology Co Ltd &Ningbo Wenda Pharma
ABBV-075 NSCLC Phase I NCT02391480 ABBV-075 128 Completed AbbVie
AZD5153 Malignant solid tumors Phase I NCT03205176 AZD5153 ± olaparib 49 Completed AstraZeneca
PLX2853 Solid tumor Phase I NCT03297424 PLX2853 49 Completed Opna-IO LLC
SYHA1801 Advanced solid tumors Phase I NCT04309968 SYHA1801 186 Recruiting CSPC ZhongQi Pharmaceutical Technology Co., Ltd.
HH-3806 Solid tumor Phase I ACTRN12622001339741 HH-3806 36 Recruiting Tigermed Australia Pty Ltd
Tigermed Australia Pty Ltd
BET ZEN-3694 Advanced and refractory solid tumors Phase Ⅰ/Ⅱ NCT05053971 ZEN-3694 ± entinostat 30 Recruiting National Cancer Institute (NCI)
NUV-868 Advanced solid tumors Phase Ⅰ/Ⅱ NCT05252390 NUV-868 ± olaparib/enzalutamide 657 Recruiting Nuvation Bio Inc.
ODM-207 Solid tumor Phase Ⅰ/Ⅱ NCT03035591 ODM-207 36 Completed Orion Corporation, Orion Pharma
Molibresib Solid tumor Phase Ⅰ/Ⅱ EUCTR2014-004982-25-ES Molibresib 225 Not yet recruiting GlaxoSmithKline, S.A.
BI-894999 Advanced solid tumors Phase I EUCTR2015-001111-12-BE BI 894999 158 Not yet recruiting SCS Boehringer Ingelheim Comm.V
MIR-34a MRX-34 NSCLC Phase I NCT01829971 MRX34 152 Terminated Mirna Therapeutics, Inc.
HDAC2 HG146 Solid tumor Phase I NCT04977167 HG146 ± PD-(L)1 96 Recruiting HitGen Inc.
Mocetinostat Advanced solid tumors Phase I NCT00323934 Mocetinostat 42 Completed Mirati Therapeutics Inc.
Pracinostat Advanced solid tumors Phase I NCT00741234 Pracinostat 85 Completed S∗BIO
SOX2 STEMVAC Stage IV non-squamous non-small cell lung cancer Phase Ⅱ NCT05242965 STEMVAC + sargramostim 40 Recruiting University of Washington
LSD1 Pulrodemstat Squamous non-small cell lung cancer Phase Ⅱ EUCTR2019-004194-95-ES Pulrodemstat + nivolumab 135 Unknown Celgene Corporation
INCB059872 Solid tumors Phase Ⅰ/Ⅱ NCT02712905 INCB059872 ± all-trans retinoic acid/azacitidine/nivolumab 116 Terminated Incyte Corporation
JBI-802 Locally advanced and metastatic solid tumor Phase Ⅰ/Ⅱ NCT05268666 JBI-802 126 Recruiting Jubilant Therapeutics Inc.
Baicalin NSCLC Phase I ChiCTR2100051276 Baicalin ± PD-1 152 Recruiting Beijing Friendship hospital, Capital Medical University
Pulrodemstat Relapsed and/or refractory solid tumors Phase I NCT02875223 Pulrodemstat + rifampicin/itraconazole 91 Not yet recruiting Celgene
Seclidemstat Advanced solid tumors Phase I NCT03895684 Seclidemstat 23 Completed Salarius Pharmaceuticals, LLC

Notes: The drug information and data in the table were sourced from Pharm Snap, ClinicalTrials.gov, EU Clinical Trials Register, Chinese Clinical Trial Registry. NA, not available; NR, not reached; ORR, objective response rate; OS, overall survival; PFS, progression-free survival; NSCLC, non-small cell lung cancer; PD-L1, programmed cell death ligand 1; PD-1, programmed cell death protein 1; EZH2, enhancer of zeste homolog 2; BRD4, bromodomain containing 4; BET, bromodomain and extraterminal; MIR-34a, microRNA 34a; HDAC2, histone deacetylase 2; SOX2, SRY-box transcription factor 2; LSD1, lysine-specific demethylase 1.

BRD4

BRD4 (bromodomain containing 4) belongs to the BET (bromodomain and extraterminal) protein family, participating in multiple biological processes including transcriptional regulation, DNA damage repair, activation of immune checkpoints, and maintenance of telomeric homeostasis.138,139 Research has shown that inhibiting the activity of BRD4 can modulate the DNA damage response, thereby increasing the sensitivity of cancer cells to stress-inducing agents.140 BET inhibitors exert a synergistic effect in combination with homologous recombination defects and PARP (poly(ADP-ribose) polymerase) inhibitors.141 Owing to its high expression in LUAD, which correlates with the malignancy of cancer cells and poor prognosis,142 BRD4 is considered a potential therapeutic target. Currently, BET protein inhibitors are still in the development and clinical trial stages, including compounds like JQ1, selective BRD2/3/4 inhibitor OTX015 (also known as MK-8628), and novel BET inhibitors like ZEN-3694.143 JQ1 demonstrates the potential to overcome cancer cell resistance, such as enhancing sensitivity in platinum-resistant cells and inhibiting the growth of BRAF-mutant cancer cells.144,145 In addition to inhibiting the functions of target proteins, Winter et al. also designed a BET protein degrader called dBET1. Treatment with dBET1 leads to the significant destabilization of the BRD4 protein, down-regulation of MYC, and inhibition of proliferation in cancer cells within the tumor.146

MIR-34a

MIR-34a (microRNA 34a) is a tumor-suppressive microRNA molecule, the expression of which is typically significantly reduced in LUAD tissues.147 Research indicates that MIR-34a, by targeting EGFR, can inhibit the growth of lung tumors,148 and simultaneously regulate the epithelial–mesenchymal transition of tumor cells to suppress tumor metastasis.149 This marked reduction in MIR-34a expression often predicts a shorter survival period and holds certain prognostic value.150,151 Furthermore, MIR-34a induces sensitivity of lung cancer cells to cisplatin by modulating the p53/miR-34a/MYCN signaling axis.152 Although MIR-34a demonstrates potential therapeutic value, achieving precise gene silencing in drug development is relatively challenging due to miRNAs being endogenously produced. Currently, drugs targeting miRNAs are still in clinical development, with only five drugs undergoing clinical trials. There was once a MIR-34a analogue called “MRX34” initially used for the treatment of solid tumors. Unfortunately, in a phase I clinical trial (NCT01829971), five severe adverse events occurred, with four patients succumbing to them. At last, the drug's development was halted in 2016.

HDAC2

HDAC2 (histone deacetylase 2) is a member of the histone deacetylase family.153 Studies have found that HDAC2 is overexpressed in lung cancer tissues. It enhances cancer cell proliferation and invasion by regulating eIF5 (eukaryotic translation initiation factor 5) and eIF6, thus adversely affecting patient prognosis.154 Scientists at Harvard Medical School confirmed in a mouse model that combining an HDAC2 inhibitor with a PD-1 (programmed cell death protein 1) antibody significantly suppresses tumor growth and improves survival rates, suggesting that HDAC2 might be a new potential option for combination therapy with PD-1 inhibitors.155 Similarly, because HDAC2 can increase the expression of angiogenic factors through its deacetylation activity, inhibiting HDAC2 may reverse this anti-angiogenic drug resistance phenomenon.156 Currently, HDAC inhibitors such as vorinostat, romidepsin, belinostat, and panobinostat have received FDA approval for marketing, primarily for lymphomas and myelomas. Chidamide has been approved by the China National Medical Products Administration for the treatment of peripheral T-cell lymphoma and breast cancer. These drugs have brought new hope to cancer treatment, and HDAC2, as an important target, may provide potential options for future cancer therapies.

TET

TET (ten-eleven translocation enzymes), including TET1, TET2, and TET3, are essential enzymes involved in the regulation of the process of converting 5-methylcytosine on DNA, controlling DNA methylation levels.157 Research has shown an association between the up-regulation of TET1 mRNA and the advanced stages of lung cancer.158 The study of Qin et al. revealed that TET enzymes suppressed the malignant progression of lung epithelial cells and LUAD by inducing low methylation of key oncogenes in the Wnt signaling pathway.159 Furthermore, Wu et al. demonstrated that patients with TET1 mutations exhibited prolonged PFS and OS when undergoing immunotherapy. This may be closely related to the increased tumor mutational burden caused by TET1 mutations and the higher presence of infiltrating T lymphocytes in tumors.160 Therefore, TET mutations can be considered as independent biomarkers for predicting immunotherapy responses in LUAD. Besides, vitamin C, as a critical cofactor, enhances TET enzyme activity, thereby facilitating active DNA demethylation.161 However, to determine the optimal dosage and precise clinical efficacy of vitamin C in fully utilizing TET therapy drugs, further large-scale clinical controlled studies are necessary.

The epigenetic therapeutic targets of LUSC

NSD3

NSD3 (nuclear receptor binding SET domain protein 3) is a histone methyltransferase primarily responsible for catalyzing trimethylation processes on histones.162 In 2021, Yuan et al. published significant research finding in Nature, revealing the pivotal role of NSD3 in the development of LUSC. They confirmed in mouse and cell models that the loss of NSD3 significantly inhibits the growth of LUSC tumors.163 Studies also indicate that NSD3 can interact with BRD4, enhancing the sensitivity of lung squamous cell cancer to BET inhibitors.164 Currently, there are several NSD3 inhibitors such as BI-9321, MS9715, and SYL2158 that have demonstrated inhibitory effects in lung cancer.165 Furthermore, research based on a proteolysis-targeting chimera (PROTAC) strategy has identified small molecule NSD3 degraders (8) capable of effectively reducing NSD3 protein levels in cell models upon single-dose administration.166 These findings offer potential directions for clinical treatment of LUSC.

SOX2

SOX2 (SRY-box transcription factor 2) is a pluripotent transcription factor involved in regulating cellular self-renewal and differentiation.167 Preclinical studies have shown that inhibiting tumor growth in LUSC cell lines can be achieved through RNA interference targeting SOX2.168 Wilbertz et al. have also identified low-level amplification of SOX2 in 68% of LUSC, distinct from LUAD. Interestingly, high expression of SOX2 in LUSC is associated with increased patient survival.169 This could be attributed to the fact that SOX2 overexpression leads to significant differentiation of squamous cells,170 thus providing better prognostic outcomes for LUSC patients. Due to the absence of an active site for small molecule inhibitors targeting SOX2, specific drugs aimed at SOX2 are not currently available for clinical use. Consequently, current research primarily focuses on understanding the regulation mechanisms and biological functions of SOX2. Some studies suggest that LUSC cell lines with SOX2 amplification may exhibit dependencies on EZH2 and associations with LSD1 (lysine-specific demethylase 1).171 Therefore, potential therapeutic strategies targeting EZH2 and LSD1 could be considered for treating this subset of LUSC.

LSD1

LSD1 is a lysine-specific demethylase primarily involved in the demethylation of di-methylated/mono-methylated histone H3 lysine 4.172 Inhibiting LSD1 can promote cell differentiation, reactivate the expression of tumor suppressor genes, and effectively control tumor development.173 Studies indicate that in LUSC expressing SOX2, the levels of LSD1 are significantly elevated, and LSD1 inhibitors exhibit a high degree of selectivity against them.174 Furthermore, LSD1 inhibitors can enhance the expression of pro-inflammatory cytokines in Treg cells and facilitate CD8+ T cell infiltration, converting “cold tumors” into “hot tumors” in immunotherapy. This enhances tumor cell sensitivity to immune checkpoint inhibitors.175 Currently, numerous LSD1 inhibitors have entered clinical trials, including ORY-1001, ORY-2001, INCB059872, IMG-7289, and CC-90011.176 However, most of these trials are focused on hematological malignancies, and further research is needed to assess their clinical efficacy in solid tumors.

SETD8

SETD8 (SET domain-containing protein 8), also known as histone H4 lysine 20 methyltransferase, primarily functions in the mono-methylation modification of lysine 20 on histone H4.177 Additionally, SETD8 is involved in the repair of DNA double-strand breaks through the c-NHEJ (canonical non-homologous end joining) pathway,178 and interacts with TWIST, enhancing the invasive capabilities of tumors.179 Research has shown that miR-502 can regulate the expression of SETD8 protein, thereby promoting the survival of lung cancer patients.180 One study demonstrated that inhibiting SETD8 could suppress the growth of LUSC and enhance its sensitivity to chemotherapy drugs.181 Furthermore, another study indicated that SETD8 inhibitors could inhibit tumor angiogenesis.182 However, now only a few SETD8 inhibitors have cellular activity, such as NSC663284, BVT948, and ryuvidine.183 Accordingly, there is an urgent need for improving the structure of these lead compounds and designing a new generation of selective and efficient SETD8 inhibitors.

KMT2D

KMT2D (lysine methyltransferase 2D), also known as MLL2 (mixed lineage leukemia 2), is primarily responsible for catalyzing amino methylation of histone H3 lysine 4.184 Gene mutations or deletions of KMT2D occur in approximately 20% of LUSC patients.185 Research by Wong and colleagues, published in Cancer Cell, demonstrates KMT2D as a crucial epigenetic target in LUSC tumorigenesis.186 Through mouse models and patient-derived xenograft experiments, they discovered that loss of KMT2D made LUSC more sensitive to RTK (receptor tyrosine kinase)-RAS inhibitors. Additionally, LUSC patients with KMT2D mutations typically exhibit lower rates of recurrence-free survival.187 The loss of KMT2D extensively impairs gene signaling in super-enhancers, suppresses glycolysis, and reduces the levels of acetylated histone H3 lysine 27.188 This discovery opens up new therapeutic strategies for KMT2D-deficient lung cancer, such as using glycolysis inhibitors. Moreover, since the loss of KMT2D leads to overactivation of MEK in the RAS/MAPK pathway,189 MAPK pathway inhibitors can also be considered potential drugs for treating KMT2D mutations.

Clinical treatment disparities

Diverse origins of cells, high-risk factors such as tobacco exposure, and variations in driver genes largely determine the distinct pathogenic mechanisms between LUAD and LUSC. In the processes of chemotherapy, targeted therapy and immunotherapy, and distinct strategies and protocols for the clinical management of LUAD and LUSC are gradually emerging (Fig. 4).

Figure 4.

Figure 4

Clinical treatment timeline for lung adenocarcinoma and squamous cell carcinoma. The emergence of EGFR-TKI drugs shifted lung adenocarcinoma from conventional chemotherapy to targeted therapy. However, over the past decade, no targeted therapies have been introduced for lung squamous cell carcinoma. With the advent of the immunotherapy era, the available treatment options for squamous cell carcinoma have gradually expanded. EGFR, epidermal growth factor receptor; HER2, human epidermal growth factor receptor 2; KRAS, Kirsten rat sarcoma viral oncogene homologue; MET, mesenchymal–epithelial transition; NSCLC, non-small cell lung cancer; PD-L1, programmed cell death ligand 1; RET, rearranged during transfection; TKI, tyrosine kinase inhibitor.

Chemotherapy

Pemetrexed is a chemotherapy drug that targets thymidylate synthase and plays a significant role in first-line treatment for LUAD (Table S2). However, in a large phase III clinical trial that was double-blind and placebo-controlled, it was found that when pemetrexed was used in combination therapy for LUSC, there was no significant difference in PFS and OS between the two groups of patients.190 This difference may be attributed to the high expression of thymidylate synthase in LUSC, which reduces patients' sensitivity to folate-based drugs. Additionally, JMDB research also found that when combining pemetrexed with platinum-based drugs for treating LUAD patients, there was a significant extension in OS, surpassing the combination of gemcitabine and platinum-based drugs. Conversely, in LUSC, the combination of gemcitabine and platinum-based drugs led to a relatively longer OS, demonstrating better efficacy compared with the combination of pemetrexed and platinum-based drugs.4 The CTONG1002 study further compared the clinical efficacy of albumin-bound paclitaxel/carboplatin with gemcitabine/carboplatin and found that the albumin-bound paclitaxel group had a superior improvement in quality of life compared with the gemcitabine group.

Targeted therapy

Compared with LUAD, LUSC has fewer driver gene targets, which limits the effectiveness of targeted therapy. However, in the SQUIRE trial, the combination of necitumumab with gemcitabine and cisplatin significantly improved the OS of late-stage LUSC patients. Consequently, necitumumab has been approved for targeted therapy in LUSC with EGFR mutations.191 Bevacizumab is a monoclonal antibody targeting vascular endothelial growth factor (VEGF), but it may increase the risk of pulmonary hemorrhage. Due to the central location and susceptibility to bleeding often observed in LUSC patients, bevacizumab is contraindicated in this subgroup. For advanced non-squamous NSCLC patients, as indicated by findings from the ARIES study, both adenocarcinoma and non-adenocarcinoma patients can benefit from bevacizumab treatment.192 Moreover, adenocarcinoma patients showed slightly higher OS and PFS compared with non-adenocarcinoma patients. Unless contraindicated, the use of bevacizumab is advantageous for LUAD patients.

Immunotherapy

At present, first-line treatments for both LUAD and LUSC involve immunotherapy drugs. In comparison to adenocarcinoma, LUSC more commonly expresses PD-L1 (programmed cell death ligand 1), and immune cell infiltration, including macrophages, is more pronounced.193 LUSC exhibits a higher somatic mutation frequency and stronger immunogenicity, offering opportunities for the treatment of advanced-stage patients. Based on studies such as KEYNOTE-024, KEYNOTE-042, and KEYNOTE-407,194, 195, 196 the 2020 CSCO (Chinese Society of Clinical Oncology) guidelines recommended the use of pembrolizumab for first-line treatment of advanced LUSC. Currently, there is a broader selection of immunotherapies for LUSC. In a phase III clinical trial, GEMSTONE-302, which included both LUSC and LUAD, it was found that the addition of avelumab to chemotherapy showed superiority in treating LUSC (hazard ratio: 0.59 vs. 0.34),197 Furthermore, on October 31, 2022, toripalimab gained approval for first-line treatment in combination with carboplatin and albumin-bound paclitaxel for locally advanced or metastatic squamous NSCLC patients. Similarly, the domestically developed immune drug camrelizumab in combination with chemotherapy is recommended in the 2022 CSCO guidelines for first-line treatment of advanced LUSC.

Conclusion and prospects

LUSC and LUAD, two subtypes of NSCLC, exhibit numerous differences in driver genes, treatment targets, and clinical efficacy. In general, gene mutations in LUAD, such as EGFR, MET, and BRAF mutations, often occur on chromosome 7, while gene mutations in LUSC, such as PIK3CA, SOX2, and TP63, are frequently observed on chromosome 3. LUAD exhibits a higher frequency of proto-oncogene mutations, particularly mutations in receptor tyrosine kinases such as EGFR, ALK, ROS, RET, MET, and HER2. In contrast, LUSC demonstrates widespread inactivation of tumor suppressor genes such as TP53, KEAP1, PTEN, and CDKN2A, along with fewer directly targeted driver genes, characteristic of a multi-driver mutation profile. Although high-frequency gene mutations in LUAD, such as EGFR and KRAS, can also be found in LUSC, their mutation frequencies are all below 5%.198 Additionally, EGFR mutations in LUSC are often not the classic sensitive mutations, leading to suboptimal responses when patients are treated with EGFR-TKIs.199 Beyond differences in gene mutation frequencies, the roles of the same genes (e.g.NOTCH1) may be entirely opposite in LUAD and LUSC (Table S1). However, such distinctions are not fully reflected in guidelines both domestically and internationally. Extensive clinical practice has shown that LUAD patients may undergo adenosquamous transformation following chemotherapy, targeted therapy, and immunotherapy.200 This suggests that adenosquamous transformation may represent a potential mechanism for the development of resistance in clinical lung cancer patients.

As one of the most crucial tumor suppressor genes in humans, TP53 carries profound clinical importance for the development of targeted therapies. Mutations in TP53 increase chromosomal instability in tumor cells, further leading to the amplification of oncogenes and loss of tumor suppressor genes.123 Therefore, besides focusing on directly targeted genes, greater emphasis should be placed on therapeutic strategies aimed at mutations in tumor suppressor genes. Despite facing challenges like drug resistance, discrepancies in animal models, and the constraints of using single-agent TP53 therapy,201 no TP53-targeted drugs have yet received approval. Nonetheless, we unanimously agree that further exploration of the biological characteristics of TP53 is highly necessary.

The discovery of epigenetic therapy targets in LUAD and LUSC, exemplified by EZH2 and NSD3, has further advanced the development of precision medicine. However, it also brings about some new challenges, such as the lack of small molecule binding sites that are difficult to drug, limited cellular activity inhibitory compounds, and potential toxicity issues. Emerging therapeutic approaches, such as the introduction of PROTACs,202 seem to offer new hope for these challenging druggable targets. Additionally, PROTACs, by virtue of their protein degradation mechanism, hold the potential to overcome the issue of small molecule drug resistance. The improvement of lead compound structures through computer molecular screening techniques may contribute to enhancing the anti-tumor activity and safety of such drugs.

Since the introduction of the first-generation EGFR-TKI drug gefitinib in the year 2000, there has been a profound transformation in the treatment landscape for LUAD and LUSC. The highly complex genomic landscape and carcinogenic pathways of LUSC contribute to its elevated mutational burden,123 posing challenges in elucidating its true driver genes. The recurrent molecular alterations in LUSC make it challenging to establish representative mouse models, greatly limiting the development and application of targeted therapies. Additionally, the higher proportion of driver gene amplifications in LUSC often results in “cross-reactivity” of PIK3CA inhibitors and FGFR1 inhibitors with wild-type genes, leading to significant toxicity reactions in clinical trials. Due to the poor selectivity of biomarkers for targeted therapy in LUSC,203 this further exacerbates the dilemma of balancing moderate benefits and high toxicity in targeted therapy for LUSC. Treatment options for LUSC are fewer compared with LUAD, and patients with advanced LUSC often have a poorer prognosis. Developing multi-targeted inhibitors targeting the characteristic multiple driver mutations of LUSC may lead to greater breakthroughs in its treatment. Owing to the specificity of tumor suppressor genes, the landscape of targeted therapy for LUSC may further focus on epigenetic therapy. The emergence of immune drugs such as PD-1/L1 inhibitors has revolutionized the treatment of LUSC, significantly improving patient survival prognosis. Therefore, future research may need to stratify management among patients treated with frontline targeted and immune therapies to determine the priority of targeted drugs.

Currently, targeting epigenetic modifications has become a crucial strategy in overcoming resistance to chemotherapy in cancer. The interplay between immunotherapy and epigenetic processes has facilitated the combination of epigenetic therapy and immunotherapy. For instance, LSD1 inhibitor enhances the immunogenicity of tumors, activates T-cell immune activity, and augments the effectiveness of immune checkpoint inhibitors.175 Additionally, the inactivation of relevant oncogenic pathways in LUSC can influence the tumor's immune microenvironment. For instance, the PI3KCA inhibitor AMG319 can lead to the activation of CD4+ and CD8+ T-cell responses and a weakening of the function of tumor-infiltrating Tregs.204 Epigenetic drugs appear to synergize with targeted therapies as well, such as the combination of histone deacetylase inhibitors with TKIs, which can restore sensitivity to targeted treatments.205 This provides a new perspective for addressing the current resistance to TKI drugs in LUAD. Based on the molecular biological characteristics of the two subtypes of lung cancer, a rational combination of epigenetic therapy, targeted therapy, and immunotherapy may achieve more effective treatment. The genetic characteristics of LUAD and LUSC allow patients to be molecularly classified based on histological classification, which will facilitate the formulation of personalized treatment plans, thereby leading to better treatment outcomes and clinical prognosis.

Conflict of interests

All authors declared no conflict of interests related to this work.

Acknowledgment

This work was supported by the Hunan Provincial Natural Science Foundation of China (No. 2023JJ50013), the Department of Science and Technology of Hunan Province, China (No. 2022ZK4052), and the Independent Exploration and Innovation Project for Graduate Students of Central South University(No. 2024ZZTS0286).

Footnotes

Peer review under responsibility of Chongqing Medical University.

Appendix A

Supplementary data to this article can be found online at https://doi.org/10.1016/j.gendis.2024.101374.

Appendix A. Supplementary data

The following is the Supplementary data to this article:

Multimedia component 1
mmc1.docx (51.3KB, docx)

References

  • 1.Siegel R.L., Giaquinto A.N., Jemal A., Cancer statistics Ca - Cancer J Clin. 2024;74(1):12–49. doi: 10.3322/caac.21820. 2024 [published correction appears in CA Cancer J Clin. 2024;74(2):203.] [DOI] [PubMed] [Google Scholar]
  • 2.Nicholson A.G., Tsao M.S., Beasley M.B., et al. The 2021 WHO classification of lung tumors: impact of advances since 2015. J Thorac Oncol. 2022;17(3):362–387. doi: 10.1016/j.jtho.2021.11.003. [DOI] [PubMed] [Google Scholar]
  • 3.Osmani L., Askin F., Gabrielson E., Li Q.K. Current WHO guidelines and the critical role of immunohistochemical markers in the subclassification of non-small cell lung carcinoma (NSCLC): Moving from targeted therapy to immunotherapy. Semin Cancer Biol. 2018;52(Pt 1):103–109. doi: 10.1016/j.semcancer.2017.11.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Scagliotti G.V., 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(21):3543–3551. doi: 10.1200/JCO.2007.15.0375. [DOI] [PubMed] [Google Scholar]
  • 5.Yuan S., Yu S.L., Chen H.Y., et al. Clustered genomic alterations in chromosome 7p dictate outcomes and targeted treatment responses of lung adenocarcinoma with EGFR-activating mutations. J Clin Oncol. 2011;29(25):3435–3442. doi: 10.1200/JCO.2011.35.3979. [DOI] [PubMed] [Google Scholar]
  • 6.Liu Y., Yin N., Wang X., et al. Chromosome 3q26 gain is an early event driving coordinated overexpression of the PRKCI, SOX2, and ECT2 oncogenes in lung squamous cell carcinoma. Cell Rep. 2020;30(3):771–782.e6. doi: 10.1016/j.celrep.2019.12.071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Tsiambas E., Mastronikolis N.S., Lefas A.Y., et al. Chromosome 7 multiplication in EGFR-positive lung carcinomas based on tissue microarray analysis. In Vivo. 2017;31(4):641–648. doi: 10.21873/invivo.11106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Shi Y., Au J.S.K., Thongprasert S., et al. A prospective, molecular epidemiology study of EGFR mutations in Asian patients with advanced non-small-cell lung cancer of adenocarcinoma histology (PIONEER) J Thorac Oncol. 2014;9(2):154–162. doi: 10.1097/JTO.0000000000000033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Chang H., Yang Y., Lee J.S., Jheon S.H., Kim Y.J., Chung J.H. Epidermal growth factor receptor gene amplification predicts worse outcome in patients with surgically resected nonadenocarcinoma lung cancer. Clin Lung Cancer. 2019;20(1):7–12.e1. doi: 10.1016/j.cllc.2018.06.003. [DOI] [PubMed] [Google Scholar]
  • 10.Peng D., Liang P., Zhong C., et al. Effect of EGFR amplification on the prognosis of EGFR-mutated advanced non-small-cell lung cancer patients: a prospective observational study. BMC Cancer. 2022;22(1):1323. doi: 10.1186/s12885-022-10390-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Lv C., Fang W., Wu N., et al. Osimertinib as neoadjuvant therapy in patients with EGFR-mutant resectable stage II-IIIB lung adenocarcinoma (NEOS): a multicenter, single-arm, open-label phase 2b trial. Lung Cancer. 2023;178:151–156. doi: 10.1016/j.lungcan.2023.02.011. [DOI] [PubMed] [Google Scholar]
  • 12.Tan A.C., Tan D.S.W. Targeted therapies for lung cancer patients with oncogenic driver molecular alterations. J Clin Oncol. 2022;40(6):611–625. doi: 10.1200/JCO.21.01626. [DOI] [PubMed] [Google Scholar]
  • 13.Zhao Y., Liu J., Cai X., et al. Efficacy and safety of first line treatments for patients with advanced epidermal growth factor receptor mutated, non-small cell lung cancer: systematic review and network meta-analysis. BMJ. 2019 doi: 10.1136/bmj.l5460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Rekowska A., Rola P., Wójcik-Superczyńska M., Chmielewska I., Krawczyk P., Milanowski J. Efficacy of osimertinib in lung squamous cell carcinoma patients with EGFR gene mutation-case report and a literature review. Curr Oncol. 2022;29(5):3531–3539. doi: 10.3390/curroncol29050285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Dong R.F., Zhu M.L., Liu M.M., et al. EGFR mutation mediates resistance to EGFR tyrosine kinase inhibitors in NSCLC: from molecular mechanisms to clinical research. Pharmacol Res. 2021;167 doi: 10.1016/j.phrs.2021.105583. [DOI] [PubMed] [Google Scholar]
  • 16.Janakiraman M., Vakiani E., Zeng Z., et al. Genomic and biological characterization of exon 4 KRAS mutations in human cancer. Cancer Res. 2010;70(14):5901–5911. doi: 10.1158/0008-5472.CAN-10-0192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Hofmann M.H., Gerlach D., Misale S., Petronczki M., Kraut N. Expanding the reach of precision oncology by drugging all KRAS mutants. Cancer Discov. 2022;12(4):924–937. doi: 10.1158/2159-8290.CD-21-1331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Prior I.A., Lewis P.D., Mattos C. A comprehensive survey of Ras mutations in cancer. Cancer Res. 2012;72(10):2457–2467. doi: 10.1158/0008-5472.CAN-11-2612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Wang H., Chi L., Yu F., et al. Annual review of KRAS inhibitors in 2022. Eur J Med Chem. 2023;249 doi: 10.1016/j.ejmech.2023.115124. [DOI] [PubMed] [Google Scholar]
  • 20.Skoulidis F., Li B.T., Dy G.K., et al. Sotorasib for lung cancers with KRAS p.G12C mutation. N Engl J Med. 2021;384(25):2371–2381. doi: 10.1056/NEJMoa2103695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Jänne P.A., Riely G.J., Gadgeel S.M., et al. Adagrasib in non-small-cell lung cancer harboring a KRAS G12C mutation. N Engl J Med. 2022;387(2):120–131. doi: 10.1056/NEJMoa2204619. [DOI] [PubMed] [Google Scholar]
  • 22.Tong X., Patel A.S., Kim E., et al. Adeno-to-squamous transition drives resistance to KRAS inhibition in LKB1 mutant lung cancer. Cancer Cell. 2024;42(3):413–428.e7. doi: 10.1016/j.ccell.2024.01.012. [DOI] [PubMed] [Google Scholar]
  • 23.Bond M.J., Chu L., Nalawansha D.A., Li K., Crews C.M. Targeted degradation of oncogenic KRASG12C by VHL-recruiting PROTACs. ACS Cent Sci. 2020;6(8):1367–1375. doi: 10.1021/acscentsci.0c00411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Yang N., Fan Z., Sun S., et al. Discovery of highly potent and selective KRASG12C degraders by VHL-recruiting PROTACs for the treatment of tumors with KRASG12C-mutation. Eur J Med Chem. 2023;261 doi: 10.1016/j.ejmech.2023.115857. [DOI] [PubMed] [Google Scholar]
  • 25.Xu J., Yu X., Martin T.C., et al. AKT degradation selectively inhibits the growth of PI3K/PTEN pathway-mutant cancers with wild-type KRAS and BRAF by destabilizing aurora kinase B. Cancer Discov. 2021;11(12):3064–3089. doi: 10.1158/2159-8290.CD-20-0815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Kostyrko K., Román M., Lee A.G., et al. UHRF1 is a mediator of KRAS driven oncogenesis in lung adenocarcinoma. Nat Commun. 2023;14:3966. doi: 10.1038/s41467-023-39591-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Rybarczyk-Kasiuchnicz A., Ramlau R., Stencel K. Treatment of brain metastases of non-small cell lung carcinoma. Int J Mol Sci. 2021;22(2):593. doi: 10.3390/ijms22020593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Ali S.M., Hensing T., Schrock A.B., et al. Comprehensive genomic profiling identifies a subset of crizotinib-responsive ALK-rearranged non-small cell lung cancer not detected by fluorescence in situ hybridization. Oncol. 2016;21(6):762–770. doi: 10.1634/theoncologist.2015-0497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Inamura K., Takeuchi K., Togashi Y., et al. EML4-ALK lung cancers are characterized by rare other mutations, a TTF-1 cell lineage, an acinar histology, and young onset. Mod Pathol. 2009;22(4):508–515. doi: 10.1038/modpathol.2009.2. [DOI] [PubMed] [Google Scholar]
  • 30.Solomon B.J., Mok T., Kim D.W., et al. First-line crizotinib versus chemotherapy in ALK-positive lung cancer. N Engl J Med. 2014;371(23):2167–2177. doi: 10.1056/NEJMoa1408440. [DOI] [PubMed] [Google Scholar]
  • 31.Solomon B.J., Bauer T.M., Mok T.S.K., et al. Efficacy and safety of first-line lorlatinib versus crizotinib in patients with advanced, ALK-positive non-small-cell lung cancer: updated analysis of data from the phase 3, randomised, open-label CROWN study. Lancet Respir Med. 2023;11(4):354–366. doi: 10.1016/S2213-2600(22)00437-4. [DOI] [PubMed] [Google Scholar]
  • 32.Shaw A.T., Solomon B.J., Besse B., et al. ALK resistance mutations and efficacy of lorlatinib in advanced anaplastic lymphoma kinase-positive non-small-cell lung cancer. J Clin Oncol. 2019;37(16):1370–1379. doi: 10.1200/JCO.18.02236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Felip E., Shaw A.T., Bearz A., et al. Intracranial and extracranial efficacy of lorlatinib in patients with ALK-positive non-small-cell lung cancer previously treated with second-generation ALK TKIs. Ann Oncol. 2021;32(5):620–630. doi: 10.1016/j.annonc.2021.02.012. [DOI] [PubMed] [Google Scholar]
  • 34.Bergethon K., Shaw A.T., Ou S.H.I., et al. ROS1 rearrangements define a unique molecular class of lung cancers. J Clin Oncol. 2012;30(8):863–870. doi: 10.1200/JCO.2011.35.6345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Drilon A., Jenkins C., Iyer S., Schoenfeld A., Keddy C., Davare M.A. ROS1-dependent cancers — biology, diagnostics and therapeutics. Nat Rev Clin Oncol. 2021;18(1):35–55. doi: 10.1038/s41571-020-0408-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Shaw A.T., Riely G.J., Bang Y.J., et al. Crizotinib in ROS1-rearranged advanced non-small-cell lung cancer (NSCLC): updated results, including overall survival, from PROFILE 1001. Ann Oncol. 2019;30(7):1121–1126. doi: 10.1093/annonc/mdz131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Chiari R., Ricciuti B., Landi L., et al. ROS1-rearranged Non-small-cell lung cancer is associated with a high rate of venous thromboembolism: analysis from a phase II, prospective, multicenter, two-arms trial (METROS) Clin Lung Cancer. 2020;21(1):15–20. doi: 10.1016/j.cllc.2019.06.012. [DOI] [PubMed] [Google Scholar]
  • 38.Alexander M., Pavlakis N., John T., et al. A multicenter study of thromboembolic events among patients diagnosed with ROS1-rearranged non-small cell lung cancer. Lung Cancer. 2020;142:34–40. doi: 10.1016/j.lungcan.2020.01.017. [DOI] [PubMed] [Google Scholar]
  • 39.Jiang Q., Li M., Li H., Chen L. Entrectinib, a new multi-target inhibitor for cancer therapy. Biomed Pharmacother. 2022;150 doi: 10.1016/j.biopha.2022.112974. [DOI] [PubMed] [Google Scholar]
  • 40.Servetto A., Esposito D., Ferrara R., et al. RET rearrangements in non-small cell lung cancer: evolving treatment landscape and future challenges. Biochim Biophys Acta Rev Cancer. 2022;1877(6) doi: 10.1016/j.bbcan.2022.188810. [DOI] [PubMed] [Google Scholar]
  • 41.Digumarthy S.R., Mendoza D.P., Lin J.J., et al. Imaging features and patterns of metastasis in non-small cell lung cancer with RET rearrangements. Cancers. 2020;12(3):693. doi: 10.3390/cancers12030693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Wang H., Wang Z., Zhang G., et al. Driver genes as predictive indicators of brain metastasis in patients with advanced NSCLC: EGFR, ALK, and RET gene mutations. Cancer Med. 2020;9(2):487–495. doi: 10.1002/cam4.2706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Takeda M., Sakai K., Nishio K., Nakagawa K. Successful long-term treatment of non-small cell lung cancer positive for RET rearrangement with pemetrexed. OncoTargets Ther. 2019;12:5355–5358. doi: 10.2147/OTT.S211582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Shih J.Y., Inoue A., Cheng R., Varea R., Kim S.W. Does pemetrexed work in targetable, nonsquamous non-small-cell lung cancer? A narrative review. Cancers. 2020;12(9):2658. doi: 10.3390/cancers12092658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Piotrowska Z., Isozaki H., Lennerz J.K., et al. Landscape of acquired resistance to osimertinib in EGFR-mutant NSCLC and clinical validation of combined EGFR and RET inhibition with osimertinib and BLU-667 for acquired RET fusion. Cancer Discov. 2018;8(12):1529–1539. doi: 10.1158/2159-8290.CD-18-1022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Rotow J., Patel J.D., Hanley M.P., et al. Osimertinib and selpercatinib efficacy, safety, and resistance in a multicenter, prospectively treated cohort of EGFR-mutant and RET fusion-positive lung cancers. Clin Cancer Res. 2023;29(16):2979–2987. doi: 10.1158/1078-0432.CCR-22-2189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Drilon A., Oxnard G., Tan D., et al. Efficacy of selpercatinib in RET fusion-positive non-small-cell lung cancer. N Engl J Med. 2020;383(9):813–824. doi: 10.1056/NEJMoa2005653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Griesinger F., Curigliano G., Thomas M., et al. Safety and efficacy of pralsetinib in RET fusion-positive non-small-cell lung cancer including as first-line therapy: update from the ARROW trial. Ann Oncol. 2022;33(11):1168–1178. doi: 10.1016/j.annonc.2022.08.002. [DOI] [PubMed] [Google Scholar]
  • 49.Wu Y.L., 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(11):1132–1143. doi: 10.1016/S2213-2600(20)30154-5. [DOI] [PubMed] [Google Scholar]
  • 50.Drilon A., Clark J.W., Weiss J., et al. Antitumor activity of crizotinib in lung cancers harboring a MET exon 14 alteration. Nat Med. 2020;26(1):47–51. doi: 10.1038/s41591-019-0716-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Awad M.M., Oxnard G.R., Jackman D.M., et al. MET exon 14 mutations in non-small-cell lung cancer are associated with advanced age and stage-dependent MET genomic amplification and c-Met overexpression. J Clin Oncol. 2016;34(7):721–730. doi: 10.1200/JCO.2015.63.4600. [DOI] [PubMed] [Google Scholar]
  • 52.Drusbosky L.M., Dawar R., Rodriguez E., Ikpeazu C.V. Therapeutic strategies in METex14 skipping mutated non-small cell lung cancer. J Hematol Oncol. 2021;14(1):129. doi: 10.1186/s13045-021-01138-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Roper N., Brown A.L., Wei J.S., et al. Clonal evolution and heterogeneity of osimertinib acquired resistance mechanisms in EGFR mutant lung cancer. Cell Rep Med. 2020;1(1) doi: 10.1016/j.xcrm.2020.100007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Wang Q., Yang S., Wang K., Sun S.Y. MET inhibitors for targeted therapy of EGFR TKI-resistant lung cancer. J Hematol Oncol. 2019;12(1):63. doi: 10.1186/s13045-019-0759-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Yu H.A., Goldberg S.B., Le X., et al. Biomarker-directed phase II platform study in patients with EGFR sensitizing mutation-positive advanced/metastatic non-small cell lung cancer whose disease has progressed on first-line osimertinib therapy (ORCHARD) Clin Lung Cancer. 2021;22(6):601–606. doi: 10.1016/j.cllc.2021.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Dagogo-Jack I., Moonsamy P., Gainor J.F., et al. A phase 2 study of capmatinib in patients with MET-altered lung cancer previously treated with a MET inhibitor. J Thorac Oncol. 2021;16(5):850–859. doi: 10.1016/j.jtho.2021.01.1605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Pearson G., Robinson F., Beers Gibson T., et al. Mitogen-activated protein (MAP) kinase pathways: regulation and physiological functions. Endocr Rev. 2001;22(2):153–183. doi: 10.1210/edrv.22.2.0428. [DOI] [PubMed] [Google Scholar]
  • 58.Papanikolaou V., Kyrodimos E., Mastronikolis N., et al. Anti-EGFR/BRAF-tyrosine kinase inhibitors in thyroid carcinoma. Cancer Diagn Progn. 2023;3(2):151–156. doi: 10.21873/cdp.10194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Leonetti A., Facchinetti F., Rossi G., et al. BRAF in non-small cell lung cancer (NSCLC): pickaxing another brick in the wall. Cancer Treat Rev. 2018;66:82–94. doi: 10.1016/j.ctrv.2018.04.006. [DOI] [PubMed] [Google Scholar]
  • 60.Marchetti A., Felicioni L., Malatesta S., et al. Clinical features and outcome of patients with non-small-cell lung cancer harboring BRAF mutations. J Clin Oncol. 2011;29(26):3574–3579. doi: 10.1200/JCO.2011.35.9638. [DOI] [PubMed] [Google Scholar]
  • 61.Lin Q., Zhang H., Ding H., et al. The association between BRAF mutation class and clinical features in BRAF-mutant Chinese non-small cell lung cancer patients. J Transl Med. 2019;17(1):298. doi: 10.1186/s12967-019-2036-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Yan N., Guo S., Zhang H., Zhang Z., Shen S., Li X. BRAF-mutated non-small cell lung cancer: current treatment status and future perspective. Front Oncol. 2022;12 doi: 10.3389/fonc.2022.863043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Dudnik E., Peled N., Nechushtan H., et al. BRAF mutant lung cancer: programmed death ligand 1 expression, tumor mutational burden, microsatellite instability status, and response to immune check-point inhibitors. J Thorac Oncol. 2018;13(8):1128–1137. doi: 10.1016/j.jtho.2018.04.024. [DOI] [PubMed] [Google Scholar]
  • 64.Mazières J., Drilon A., Lusque A., et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: results from the IMMUNOTARGET registry. Ann Oncol. 2019;30(8):1321–1328. doi: 10.1093/annonc/mdz167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Rihawi K., Giannarelli D., Galetta D., et al. BRAF mutant NSCLC and immune checkpoint inhibitors: results from a real-world experience. J Thorac Oncol. 2019;14(3):e57–e59. doi: 10.1016/j.jtho.2018.11.036. [DOI] [PubMed] [Google Scholar]
  • 66.Subbiah V., Baik C., Kirkwood J.M. Clinical development of BRAF plus MEK inhibitor combinations. Trends Cancer. 2020;6(9):797–810. doi: 10.1016/j.trecan.2020.05.009. [DOI] [PubMed] [Google Scholar]
  • 67.Planchard D., Besse B., Groen H.J.M., et al. Phase 2 study of dabrafenib plus trametinib in patients with BRAF V600E-mutant metastatic NSCLC: updated 5-year survival rates and genomic analysis. J Thorac Oncol. 2022;17(1):103–115. doi: 10.1016/j.jtho.2021.08.011. [DOI] [PubMed] [Google Scholar]
  • 68.Auliac J.B., Bayle S., Do P., et al. Efficacy of dabrafenib plus trametinib combination in patients with BRAF V600E-mutant NSCLC in real-world setting: gfpc 01-2019. Cancers. 2020;12(12):3608. doi: 10.3390/cancers12123608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Brazel D., Kroening G., Nagasaka M. Non-small cell lung cancer with EGFR or HER2 exon 20 insertion mutations: diagnosis and treatment options. BioDrugs. 2022;36(6):717–729. doi: 10.1007/s40259-022-00556-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Tomizawa K., Suda K., Onozato R., et al. Prognostic and predictive implications of HER2/ERBB2/neu gene mutations in lung cancers. Lung Cancer. 2011;74(1):139–144. doi: 10.1016/j.lungcan.2011.01.014. [DOI] [PubMed] [Google Scholar]
  • 71.Offin M., Feldman D., Ni A., et al. Frequency and outcomes of brain metastases in patients with HER2-mutant lung cancers. Cancer. 2019;125(24):4380–4387. doi: 10.1002/cncr.32461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Li B.T., Smit E.F., Goto Y., et al. Trastuzumab deruxtecan in HER2-mutant non-small-cell lung cancer. N Engl J Med. 2022;386(3):241–251. doi: 10.1056/NEJMoa2112431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Ettinger D.S., Wood D.E., Aisner D.L., et al. Non-small cell lung cancer, version 3.2022, NCCN clinical practice guidelines in oncology. J Natl Compr Cancer Netw. 2022;20(5):497–530. doi: 10.6004/jnccn.2022.0025. [DOI] [PubMed] [Google Scholar]
  • 74.Song Z., Li Y., Chen S., et al. Efficacy and safety of pyrotinib in advanced lung adenocarcinoma with HER2 mutations: a multicenter, single-arm, phase II trial. BMC Med. 2022;20(1):42. doi: 10.1186/s12916-022-02245-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Paik P.K., Shen R., Won H., et al. Next-generation sequencing of stage IV squamous cell lung cancers reveals an association of PI3K aberrations and evidence of clonal heterogeneity in patients with brain metastases. Cancer Discov. 2015;5(6):610–621. doi: 10.1158/2159-8290.CD-14-1129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Pu X., Hildebrandt M.A.T., Lu C., et al. PI3K/PTEN/AKT/mTOR pathway genetic variation predicts toxicity and distant progression in lung cancer patients receiving platinum-based chemotherapy. Lung Cancer. 2011;71(1):82–88. doi: 10.1016/j.lungcan.2010.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Dutt A., Ramos A.H., Hammerman P.S., et al. Inhibitor-sensitive FGFR1 amplification in human non-small cell lung cancer. PLoS One. 2011;6(6) doi: 10.1371/journal.pone.0020351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Mason I. Initiation to end point: the multiple roles of fibroblast growth factors in neural development. Nat Rev Neurosci. 2007;8(8):583–596. doi: 10.1038/nrn2189. [DOI] [PubMed] [Google Scholar]
  • 79.Weiss J., Sos M.L., Seidel D., et al. Frequent and focal FGFR1 amplification associates with therapeutically tractable FGFR1 dependency in squamous cell lung cancer. Sci Transl Med. 2010;2(62) doi: 10.1126/scitranslmed.3001451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Jiang T., Gao G., Fan G., Li M., Zhou C. FGFR1 amplification in lung squamous cell carcinoma: a systematic review with meta-analysis. Lung Cancer. 2015;87(1):1–7. doi: 10.1016/j.lungcan.2014.11.009. [DOI] [PubMed] [Google Scholar]
  • 81.Paik P.K., Shen R., Berger M.F., et al. A phase Ib open-label multicenter study of AZD4547 in patients with advanced squamous cell lung cancers. Clin Cancer Res. 2017;23(18):5366–5373. doi: 10.1158/1078-0432.CCR-17-0645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Aggarwal C., Redman M.W., Lara P.N., Jr., et al. SWOG S1400D ( NCT02965378), a phase II study of the fibroblast growth factor receptor inhibitor AZD4547 in previously treated patients with fibroblast growth factor pathway-activated stage IV squamous cell lung cancer (lung-MAP substudy) J Thorac Oncol. 2019;14(10):1847–1852. doi: 10.1016/j.jtho.2019.05.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Addeo A., Rothschild S.I., Holer L., et al. Fibroblast growth factor receptor (FGFR) inhibitor rogaratinib in patients with advanced pretreated squamous-cell non-small cell lung cancer over-expressing FGFR mRNA: the SAKK 19/18 phase II study. Lung Cancer. 2022;172:154–159. doi: 10.1016/j.lungcan.2022.08.016. [DOI] [PubMed] [Google Scholar]
  • 84.Nogova L., Sequist L.V., Garcia J.M.P., et al. Evaluation of BGJ398, a fibroblast growth factor receptor 1-3 kinase inhibitor, in patients with advanced solid tumors harboring genetic alterations in fibroblast growth factor receptors: results of a global phase I, dose-escalation and dose-expansion study. J Clin Oncol. 2017;35(2):157–165. doi: 10.1200/JCO.2016.67.2048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Elkamhawy A., Lu Q., Nada H., Woo J., Quan G., Lee K. The journey of DDR1 and DDR2 kinase inhibitors as rising stars in the fight against cancer. Int J Mol Sci. 2021;22(12):6535. doi: 10.3390/ijms22126535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Payne L.S., Huang P.H. Discoidin domain receptor 2 signaling networks and therapy in lung cancer. J Thorac Oncol. 2014;9(6):900–904. doi: 10.1097/JTO.0000000000000164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Miao L., Wang Y., Zhu S., et al. Identification of novel driver mutations of the discoidin domain receptor 2 (DDR2) gene in squamous cell lung cancer of Chinese patients. BMC Cancer. 2014;14(1):369. doi: 10.1186/1471-2407-14-369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Day E., Waters B., Spiegel K., et al. Inhibition of collagen-induced discoidin domain receptor 1 and 2 activation by imatinib, nilotinib and dasatinib. Eur J Pharmacol. 2008;599(1–3):44–53. doi: 10.1016/j.ejphar.2008.10.014. [DOI] [PubMed] [Google Scholar]
  • 89.Brunner A.M., Costa D.B., Heist R.S., et al. Treatment-related toxicities in a phase II trial of dasatinib in patients with squamous cell carcinoma of the lung. J Thorac Oncol. 2013;8(11):1434–1437. doi: 10.1097/JTO.0b013e3182a47162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Pons-Tostivint E., Lugat A., Fontenau J.F., Denis M.G., Bennouna J. STK11/LKB1 modulation of the immune response in lung cancer: from biology to therapeutic impact. Cells. 2021;10(11):3129. doi: 10.3390/cells10113129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Krishnamurthy N., Goodman A.M., Barkauskas D.A., Kurzrock R. STK11 alterations in the pan-cancer setting: prognostic and therapeutic implications. Eur J Cancer. 2021;148:215–229. doi: 10.1016/j.ejca.2021.01.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Pierce S.E., Granja J.M., Corces M.R., et al. LKB1 inactivation modulates chromatin accessibility to drive metastatic progression. Nat Cell Biol. 2021;23(8):915–924. doi: 10.1038/s41556-021-00728-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Ndembe G., Intini I., Perin E., et al. LKB1: can we target an hidden target? Focus on NSCLC. Front Oncol. 2022;12 doi: 10.3389/fonc.2022.889826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Eichner L.J., Curtis S.D., Brun S.N., et al. HDAC3 is critical in tumor development and therapeutic resistance in Kras-mutant non-small cell lung cancer. Sci Adv. 2023;9(11) doi: 10.1126/sciadv.add3243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Dempke W.C.M., Reck M. KEAP1/NRF2 (NFE2L2) mutations in NSCLC - fuel for a superresistant phenotype? Lung Cancer. 2021;159:10–17. doi: 10.1016/j.lungcan.2021.07.006. [DOI] [PubMed] [Google Scholar]
  • 96.Wohlhieter C.A., Richards A.L., Uddin F., et al. Concurrent mutations in STK11 and KEAP1 promote ferroptosis protection and SCD1 dependence in lung cancer. Cell Rep. 2020;33(9) doi: 10.1016/j.celrep.2020.108444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Galán-Cobo A., Sitthideatphaiboon P., Qu X., et al. LKB1 and KEAP1/NRF2 pathways cooperatively promote metabolic reprogramming with enhanced glutamine dependence in KRAS-mutant lung adenocarcinoma. Cancer Res. 2019;79(13):3251–3267. doi: 10.1158/0008-5472.CAN-18-3527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Koppula P., Olszewski K., Zhang Y., et al. KEAP1 deficiency drives glucose dependency and sensitizes lung cancer cells and tumors to GLUT inhibition. iScience. 2021;24(6) doi: 10.1016/j.isci.2021.102649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Shibata T., Saito S., Kokubu A., Suzuki T., Yamamoto M., Hirohashi S. Global downstream pathway analysis reveals a dependence of oncogenic NF-E2-related factor 2 mutation on the mTOR growth signaling pathway. Cancer Res. 2010;70(22):9095–9105. doi: 10.1158/0008-5472.CAN-10-0384. [DOI] [PubMed] [Google Scholar]
  • 100.Zhang Y., Williams-Villalobo A., Godavarthi J.D., Shakoor F., Xiong S., Liu B. Integrative bioinformatic analysis of p53 and pathway alterations in two different lung cancer subtypes. Biochem Biophys Rep. 2023;33 doi: 10.1016/j.bbrep.2022.101404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Schwaederle M., Elkin S.K., Tomson B.N., Carter J.L., Kurzrock R. Squamousness: next-generation sequencing reveals shared molecular features across squamous tumor types. Cell Cycle. 2015;14(14):2355–2361. doi: 10.1080/15384101.2015.1053669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Xu S., Wang Y., Ren F., et al. Impact of genetic alterations on outcomes of patients with stage I nonsmall cell lung cancer: an analysis of the cancer genome atlas data. Cancer Med. 2020;9(20):7686–7694. doi: 10.1002/cam4.3403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Gu J., Zhou Y., Huang L., et al. TP53 mutation is associated with a poor clinical outcome for non-small cell lung cancer: evidence from a meta-analysis. Mol Clin Oncol. 2016;5(6):705–713. doi: 10.3892/mco.2016.1057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Kryczka J., Kryczka J., Czarnecka-Chrebelska K.H., Brzeziańska-Lasota E. Molecular mechanisms of chemoresistance induced by cisplatin in NSCLC cancer therapy. Int J Mol Sci. 2021;22(16):8885. doi: 10.3390/ijms22168885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Wang M., Zhu L., Yang X., Li J., Liu Y.E., Tang Y. Targeting immune cell types of tumor microenvironment to overcome resistance to PD-1/PD-L1 blockade in lung cancer. Front Pharmacol. 2023;14 doi: 10.3389/fphar.2023.1132158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Abrams S.L., Duda P., Akula S.M., et al. Effects of the mutant TP53 reactivator APR-246 on therapeutic sensitivity of pancreatic cancer cells in the presence and absence of WT-TP53. Cells. 2022;11(5):794. doi: 10.3390/cells11050794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Synnott N.C., O'Connell D., Crown J., Duffy M.J. COTI-2 reactivates mutant p53 and inhibits growth of triple-negative breast cancer cells. Breast Cancer Res Treat. 2020;179(1):47–56. doi: 10.1007/s10549-019-05435-1. [DOI] [PubMed] [Google Scholar]
  • 108.Kong A., Mehanna H. WEE1 inhibitor: clinical development. Curr Oncol Rep. 2021;23(9):107. doi: 10.1007/s11912-021-01098-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Konopleva M., Martinelli G., Daver N., et al. MDM2 inhibition: an important step forward in cancer therapy. Leukemia. 2020;34(11):2858–2874. doi: 10.1038/s41375-020-0949-z. [DOI] [PubMed] [Google Scholar]
  • 110.Haupt S., Mejía-Hernández J.O., Vijayakumaran R., Keam S.P., Haupt Y. The long and the short of it: the MDM4 tail so far. J Mol Cell Biol. 2019;11(3):231–244. doi: 10.1093/jmcb/mjz007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Goel S., Bergholz J.S., Zhao J.J. Targeting CDK4 and CDK6 in cancer. Nat Rev Cancer. 2022;22(6):356–372. doi: 10.1038/s41568-022-00456-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Jin G., Kim M.J., Jeon H.S., et al. PTEN mutations and relationship to EGFR, ERBB2, KRAS, and TP53 mutations in non-small cell lung cancers. Lung Cancer. 2010;69(3):279–283. doi: 10.1016/j.lungcan.2009.11.012. [DOI] [PubMed] [Google Scholar]
  • 113.Xiao J., Hu C.P., He B.X., et al. PTEN expression is a prognostic marker for patients with non-small cell lung cancer: a systematic review and meta-analysis of the literature. Oncotarget. 2016;7(36):57832–57840. doi: 10.18632/oncotarget.11068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Spoerke J.M., O'Brien C., Huw L., et al. Phosphoinositide 3-kinase (PI3K) pathway alterations are associated with histologic subtypes and are predictive of sensitivity to PI3K inhibitors in lung cancer preclinical models. Clin Cancer Res. 2012;18(24):6771–6783. doi: 10.1158/1078-0432.CCR-12-2347. [DOI] [PubMed] [Google Scholar]
  • 115.Janku F., Yap T.A., Meric-Bernstam F. Targeting the PI3K pathway in cancer: are we making headway? Nat Rev Clin Oncol. 2018;15:273–291. doi: 10.1038/nrclinonc.2018.28. [DOI] [PubMed] [Google Scholar]
  • 116.Bergholz J.S., Wang Q., Wang Q., et al. PI3Kβ controls immune evasion in PTEN-deficient breast tumours. Nature. 2023;617(7959):139–146. doi: 10.1038/s41586-023-05940-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Zhang K., Hong X., Song Z., et al. Identification of deleterious NOTCH mutation as novel predictor to efficacious immunotherapy in NSCLC. Clin Cancer Res. 2020;26(14):3649–3661. doi: 10.1158/1078-0432.CCR-19-3976. [DOI] [PubMed] [Google Scholar]
  • 118.Sinicropi-Yao S.L., Amann J.M., Lopez D.L.Y., Cerciello F., Coombes K.R., Carbone D.P. Co-expression analysis reveals mechanisms underlying the varied roles of NOTCH1 in NSCLC. J Thorac Oncol. 2019;14(2):223–236. doi: 10.1016/j.jtho.2018.10.162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Yoshida K., Gowers K.H.C., Lee-Six H., et al. Tobacco smoking and somatic mutations in human bronchial epithelium. Nature. 2020;578(7794):266–272. doi: 10.1038/s41586-020-1961-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Zhou C., Huang D., Fan Y., et al. Tislelizumab versus docetaxel in patients with previously treated advanced NSCLC (RATIONALE-303): a phase 3, open-label, randomized controlled trial. J Thorac Oncol. 2023;18(1):93–105. doi: 10.1016/j.jtho.2022.09.217. [DOI] [PubMed] [Google Scholar]
  • 121.Licciulli S., Avila J.L., Hanlon L., et al. Notch1 is required for Kras-induced lung adenocarcinoma and controls tumor cell survival via p53. Cancer Res. 2013;73(19):5974–5984. doi: 10.1158/0008-5472.CAN-13-1384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Timofeeva M.N., Hung R.J., Rafnar T., et al. Influence of common genetic variation on lung cancer risk: meta-analysis of 14,900 cases and 29,485 controls. Hum Mol Genet. 2012;21(22):4980–4995. doi: 10.1093/hmg/dds334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Cancer Genome Atlas Research Network Comprehensive genomic characterization of squamous cell lung cancers. Nature. 2012;489(7417):519–525. doi: 10.1038/nature11404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Zhao R., Choi B.Y., Lee M.H., Bode A.M., Dong Z. Implications of genetic and epigenetic alterations of CDKN2A (p16(INK4a)) in cancer. EBioMedicine. 2016;8:30–39. doi: 10.1016/j.ebiom.2016.04.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Tong J., Sun X., Cheng H., et al. Expression of p16 in non-small cell lung cancer and its prognostic significance: a meta-analysis of published literatures. Lung Cancer. 2011;74(2):155–163. doi: 10.1016/j.lungcan.2011.04.019. [DOI] [PubMed] [Google Scholar]
  • 126.Jeong E.H., Lee T.G., Ko Y.J., et al. Anti-tumor effect of CDK inhibitors on CDKN2A-defective squamous cell lung cancer cells. Cell Oncol. 2018;41(6):663–675. doi: 10.1007/s13402-018-0404-6. [DOI] [PubMed] [Google Scholar]
  • 127.Dias e Silva D., Borba G.B., Beal J.R., et al. Response to abemaciclib and immunotherapy rechallenge with nivolumab and ipilimumab in a heavily pretreated TMB-H metastatic squamous cell lung cancer with CDKN2A mutation, PIK3CA amplification and TPS 80%: a case report. Int J Mol Sci. 2023;24(4):4209. doi: 10.3390/ijms24044209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Allis C.D., Jenuwein T. The molecular hallmarks of epigenetic control. Nat Rev Genet. 2016;17(8):487–500. doi: 10.1038/nrg.2016.59. [DOI] [PubMed] [Google Scholar]
  • 129.Hogg S.J., Beavis P.A., Dawson M.A., Johnstone R.W. Targeting the epigenetic regulation of antitumour immunity. Nat Rev Drug Discov. 2020;19(11):776–800. doi: 10.1038/s41573-020-0077-5. [DOI] [PubMed] [Google Scholar]
  • 130.Kim H.J., Cantor H., Cosmopoulos K. Overcoming immune checkpoint blockade resistance via EZH2 inhibition. Trends Immunol. 2020;41(10):948–963. doi: 10.1016/j.it.2020.08.010. [DOI] [PubMed] [Google Scholar]
  • 131.Hu F.F., Chen H., Duan Y., et al. CBX2 and EZH2 cooperatively promote the growth and metastasis of lung adenocarcinoma. Mol Ther Nucleic Acids. 2021;27:670–684. doi: 10.1016/j.omtn.2021.12.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Behrens C., Solis L.M., Lin H., et al. EZH2 protein expression associates with the early pathogenesis, tumor progression, and prognosis of non-small cell lung carcinoma. Clin Cancer Res. 2013;19(23):6556–6565. doi: 10.1158/1078-0432.CCR-12-3946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Chen Y., Liu X., Li Y., Quan C., Zheng L., Huang K. Lung cancer therapy targeting histone methylation: opportunities and challenges. Comput Struct Biotechnol J. 2018;16:211–223. doi: 10.1016/j.csbj.2018.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Fan K., Zhang C.L., Qi Y.F., et al. Prognostic value of EZH2 in non-small-cell lung cancers: a meta-analysis and bioinformatics analysis. BioMed Res Int. 2020;2020 doi: 10.1155/2020/2380124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Kim N.Y., Pyo J.S. Clinicopathological significance and prognostic role of EZH2 expression in non-small cell lung cancer. Pathol Res Pract. 2017;213(7):778–782. doi: 10.1016/j.prp.2017.04.002. [DOI] [PubMed] [Google Scholar]
  • 136.Quan C., Chen Y., Wang X., et al. Loss of histone lysine methyltransferase EZH2 confers resistance to tyrosine kinase inhibitors in non-small cell lung cancer. Cancer Lett. 2020;495:41–52. doi: 10.1016/j.canlet.2020.09.003. [DOI] [PubMed] [Google Scholar]
  • 137.Eich M.L., Athar M., Ferguson J.E., III, Varambally S. EZH2-targeted therapies in cancer: hype or a reality. Cancer Res. 2020;80(24):5449–5458. doi: 10.1158/0008-5472.CAN-20-2147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Wu S.Y., Chiang C.M. The double bromodomain-containing chromatin adaptor Brd4 and transcriptional regulation. J Biol Chem. 2007;282(18):13141–13145. doi: 10.1074/jbc.R700001200. [DOI] [PubMed] [Google Scholar]
  • 139.Donati B., Lorenzini E., Ciarrocchi A. BRD4 and Cancer: going beyond transcriptional regulation. Mol Cancer. 2018;17(1):164. doi: 10.1186/s12943-018-0915-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Zhang J., Dulak A.M., Hattersley M.M., et al. BRD4 facilitates replication stress-induced DNA damage response. Oncogene. 2018;37(28):3763–3777. doi: 10.1038/s41388-018-0194-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Sun C., Yin J., Fang Y., et al. BRD4 inhibition is synthetic lethal with PARP inhibitors through the induction of homologous recombination deficiency. Cancer Cell. 2018;33(3):401–416.e8. doi: 10.1016/j.ccell.2018.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Lesbon J.C.C., Garnica T.K., Xavier P.L.P., et al. A screening of epigenetic therapeutic targets for non-small cell lung cancer reveals PADI4 and KDM6B as promising candidates. Int J Mol Sci. 2022;23(19) doi: 10.3390/ijms231911911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Sun Y., Han J., Wang Z., Li X., Sun Y., Hu Z. Safety and efficacy of bromodomain and extra-terminal inhibitors for the treatment of hematological malignancies and solid tumors: a systematic study of clinical trials. Front Pharmacol. 2020;11 doi: 10.3389/fphar.2020.621093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Nakamura Y., Hattori N., Iida N., et al. Targeting of super-enhancers and mutant BRAF can suppress growth of BRAF-mutant colon cancer cells via repression of MAPK signaling pathway. Cancer Lett. 2017;402:100–109. doi: 10.1016/j.canlet.2017.05.017. [DOI] [PubMed] [Google Scholar]
  • 145.Bacabac M., Xu W. Oncogenic super-enhancers in cancer: mechanisms and therapeutic targets. Cancer Metastasis Rev. 2023;42(2):471–480. doi: 10.1007/s10555-023-10103-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Winter G.E., Buckley D.L., Paulk J., et al. DRUG DEVELOPMENT. Phthalimide conjugation as a strategy for in vivo target protein degradation. Science. 2015;348(6241):1376–1381. doi: 10.1126/science.aab1433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Mudduluru G., Ceppi P., Kumarswamy R., Scagliotti G.V., Papotti M., Allgayer H. Regulation of Axl receptor tyrosine kinase expression by miR-34a and miR-199a/b in solid cancer. Oncogene. 2011;30(25):2888–2899. doi: 10.1038/onc.2011.13. [DOI] [PubMed] [Google Scholar]
  • 148.Li Y.L., Liu X.M., Zhang C.Y., et al. microRNA-34a/EGFR axis plays pivotal roles in lung tumorigenesis. Oncogenesis. 2017;6(8) doi: 10.1038/oncsis.2017.50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Li X.J., Ren Z.J., Tang J.H. microRNA-34a: a potential therapeutic target in human cancer. Cell Death Dis. 2014;5(7) doi: 10.1038/cddis.2014.270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Gallardo E., Navarro A., Viñolas N., et al. miR-34a as a prognostic marker of relapse in surgically resected non-small-cell lung cancer. Carcinogenesis. 2009;30(11):1903–1909. doi: 10.1093/carcin/bgp219. [DOI] [PubMed] [Google Scholar]
  • 151.Zhong S., Golpon H., Zardo P., Borlak J. miRNAs in lung cancer. A systematic review identifies predictive and prognostic miRNA candidates for precision medicine in lung cancer. Transl Res. 2021;230:164–196. doi: 10.1016/j.trsl.2020.11.012. [DOI] [PubMed] [Google Scholar]
  • 152.Song C., Lu P., Sun G., Yang L., Wang Z., Wang Z. miR-34a sensitizes lung cancer cells to cisplatin via p53/miR-34a/MYCN axis. Biochem Biophys Res Commun. 2017;482(1):22–27. doi: 10.1016/j.bbrc.2016.11.037. [DOI] [PubMed] [Google Scholar]
  • 153.Jo H., Shim K., Kim H.U., Jung H.S., Jeoung D. HDAC2 as a target for developing anti-cancer drugs. Comput Struct Biotechnol J. 2023;21:2048–2057. doi: 10.1016/j.csbj.2023.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Cai S.X., Chen W.S., Zeng W., Cheng X.F., Lin M.B., Wang J.S. Roles of HDAC2, eIF5, and eIF6 in lung cancer tumorigenesis. Curr Med Sci. 2021;41(4):764–769. doi: 10.1007/s11596-021-2389-z. [DOI] [PubMed] [Google Scholar]
  • 155.Gao Y., Nihira N.T., Bu X., et al. Acetylation-dependent regulation of PD-L1 nuclear translocation dictates the efficacy of anti-PD-1 immunotherapy. Nat Cell Biol. 2020;22(9):1064–1075. doi: 10.1038/s41556-020-0562-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Eckschlager T., Plch J., Stiborova M., Hrabeta J. Histone deacetylase inhibitors as anticancer drugs. Int J Mol Sci. 2017;18(7):1414. doi: 10.3390/ijms18071414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Joshi K., Liu S., Breslin S.J.P., Zhang J. Mechanisms that regulate the activities of TET proteins. Cell Mol Life Sci. 2022;79(7):363. doi: 10.1007/s00018-022-04396-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Filipczak P.T., Leng S., Tellez C.S., et al. p53-suppressed oncogene TET1 prevents cellular aging in lung cancer. Cancer Res. 2019;79(8):1758–1768. doi: 10.1158/0008-5472.CAN-18-1234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Xu Q., Wang C., Zhou J.X., et al. Loss of TET reprograms Wnt signaling through impaired demethylation to promote lung cancer development. Proc Natl Acad Sci USA. 2022;119(6) doi: 10.1073/pnas.2107599119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Wu H.X., Chen Y.X., Wang Z.X., et al. Alteration in TET1 as potential biomarker for immune checkpoint blockade in multiple cancers. J Immunother Cancer. 2019;7(1):264. doi: 10.1186/s40425-019-0737-3. [published correction appears in J Immunother Cancer. 2020;8(1):e0737-3corr1.] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Yue X., Rao A. TET family dioxygenases and the TET activator vitamin C in immune responses and cancer. Blood. 2020;136(12):1394–1401. doi: 10.1182/blood.2019004158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Topchu I., Pangeni R.P., Bychkov I., et al. The role of NSD1, NSD2, and NSD3 histone methyltransferases in solid tumors. Cell Mol Life Sci. 2022;79(6):285. doi: 10.1007/s00018-022-04321-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Yuan G., Flores N.M., Hausmann S., et al. Elevated NSD3 histone methylation activity drives squamous cell lung cancer. Nature. 2021;590(7846):504–508. doi: 10.1038/s41586-020-03170-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Rahman S., Sowa M.E., Ottinger M., et al. The Brd4 extraterminal domain confers transcription activation independent of pTEFb by recruiting multiple proteins, including NSD3. Mol Cell Biol. 2011;31(13):2641–2652. doi: 10.1128/MCB.01341-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Xiu S., Chi X., Jia Z., et al. NSD3: advances in cancer therapeutic potential and inhibitors research. Eur J Med Chem. 2023;256 doi: 10.1016/j.ejmech.2023.115440. [DOI] [PubMed] [Google Scholar]
  • 166.Sun Y., Zhang Y., Chen X., et al. Discovery of a potent and selective proteolysis targeting chimera (PROTAC) degrader of NSD3 histone methyltransferase. Eur J Med Chem. 2022;239 doi: 10.1016/j.ejmech.2022.114528. [DOI] [PubMed] [Google Scholar]
  • 167.Novak D., Hüser L., Elton J.J., Umansky V., Altevogt P., Utikal J. SOX2 in development and cancer biology. Semin Cancer Biol. 2020;67(pt 1):74–82. doi: 10.1016/j.semcancer.2019.08.007. [DOI] [PubMed] [Google Scholar]
  • 168.Bass A.J., Watanabe H., Mermel C.H., et al. SOX2 is an amplified lineage-survival oncogene in lung and esophageal squamous cell carcinomas. Nat Genet. 2009;41(11):1238–1242. doi: 10.1038/ng.465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Wilbertz T., Wagner P., Petersen K., et al. SOX2 gene amplification and protein overexpression are associated with better outcome in squamous cell lung cancer. Mod Pathol. 2011;24(7):944–953. doi: 10.1038/modpathol.2011.49. [DOI] [PubMed] [Google Scholar]
  • 170.Zhang S., Xiong X., Sun Y. Functional characterization of SOX2 as an anticancer target. Signal Transduct Targeted Ther. 2020;5:135. doi: 10.1038/s41392-020-00242-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Satpathy S., Krug K., Jean Beltran P.M., et al. A proteogenomic portrait of lung squamous cell carcinoma. Cell. 2021;184(16):4348–4371.e40. doi: 10.1016/j.cell.2021.07.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Shi Y., Lan F., Matson C., et al. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell. 2004;119(7):941–953. doi: 10.1016/j.cell.2004.12.012. [DOI] [PubMed] [Google Scholar]
  • 173.Zheng Y.C., Ma J., Wang Z., et al. A systematic review of histone lysine-specific demethylase 1 and its inhibitors. Med Res Rev. 2015;35(5):1032–1071. doi: 10.1002/med.21350. [DOI] [PubMed] [Google Scholar]
  • 174.Zhang X., Lu F., Wang J., et al. Pluripotent stem cell protein Sox2 confers sensitivity to LSD1 inhibition in cancer cells. Cell Rep. 2013;5(2):445–457. doi: 10.1016/j.celrep.2013.09.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Bao L., Zhu P., Mou Y., Song Y., Qin Y. Targeting LSD1 in tumor immunotherapy: rationale, challenges and potential. Front Immunol. 2023;14 doi: 10.3389/fimmu.2023.1214675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Zhang X., Wang X., Wu T., et al. Therapeutic potential of targeting LSD1/KDM1A in cancers. Pharmacol Res. 2022;175 doi: 10.1016/j.phrs.2021.105958. [DOI] [PubMed] [Google Scholar]
  • 177.Brustel J., Tardat M., Kirsh O., Grimaud C., Julien E. Coupling mitosis to DNA replication: the emerging role of the histone H4-lysine 20 methyltransferase PR-Set7. Trends Cell Biol. 2011;21(8):452–460. doi: 10.1016/j.tcb.2011.04.006. [DOI] [PubMed] [Google Scholar]
  • 178.Dulev S., Tkach J., Lin S., Batada N.N. SET8 methyltransferase activity during the DNA double-strand break response is required for recruitment of 53BP1. EMBO Rep. 2014;15(11):1163–1174. doi: 10.15252/embr.201439434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Yang F., Sun L., Li Q., et al. SET8 promotes epithelial-mesenchymal transition and confers TWIST dual transcriptional activities. EMBO J. 2012;31(1):110–123. doi: 10.1038/emboj.2011.364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Xu J., Yin Z., Gao W., et al. Genetic variation in a microRNA-502 minding site in SET8 gene confers clinical outcome of non-small cell lung cancer in a Chinese population. PLoS One. 2013;8(10) doi: 10.1371/journal.pone.0077024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Lazarus K.A., Hadi F., Zambon E., et al. BCL11A interacts with SOX2 to control the expression of epigenetic regulators in lung squamous carcinoma. Nat Commun. 2018;9(1):3327. doi: 10.1038/s41467-018-05790-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Choi D.K., Kim Y.K., Park S.W., et al. The histone lysine methyltransferase SETD8 regulates angiogenesis through HES-1 in human umbilical vein endothelial cells. Sci Rep. 2020;10 doi: 10.1038/s41598-020-69103-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Milite C., Feoli A., Viviano M., et al. The emerging role of lysine methyltransferase SETD8 in human diseases. Clin Epigenet. 2016;8(1):102. doi: 10.1186/s13148-016-0268-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Froimchuk E., Jang Y., Ge K. Histone H3 lysine 4 methyltransferase KMT2D. Gene. 2017;627:337–342. doi: 10.1016/j.gene.2017.06.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Campbell J.D., Alexandrov A., Kim J., et al. Distinct patterns of somatic genome alterations in lung adenocarcinomas and squamous cell carcinomas. Nat Genet. 2016;48(6):607–616. doi: 10.1038/ng.3564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Pan Y., Han H., Hu H., et al. KMT2D deficiency drives lung squamous cell carcinoma and hypersensitivity to RTK-RAS inhibition. Cancer Cell. 2023;41(1):88–105.e8. doi: 10.1016/j.ccell.2022.11.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Choi M., Kadara H., Zhang J., et al. Mutation profiles in early-stage lung squamous cell carcinoma with clinical follow-up and correlation with markers of immune function. Ann Oncol. 2017;28(1):83–89. doi: 10.1093/annonc/mdw437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Alam H., Tang M., Maitituoheti M., et al. KMT2D deficiency impairs super-enhancers to confer a glycolytic vulnerability in lung cancer. Cancer Cell. 2020;37(4):599–617.e7. doi: 10.1016/j.ccell.2020.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Tsai I.C., McKnight K., McKinstry S.U., et al. Small molecule inhibition of RAS/MAPK signaling ameliorates developmental pathologies of Kabuki Syndrome. Sci Rep. 2018;8 doi: 10.1038/s41598-018-28709-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Ciuleanu T., Brodowicz T., Zielinski C., et al. Maintenance pemetrexed plus best supportive care versus placebo plus best supportive care for non-small-cell lung cancer: a randomised, double-blind, phase 3 study. Lancet. 2009;374:1432–1440. doi: 10.1016/S0140-6736(09)61497-5. [DOI] [PubMed] [Google Scholar]
  • 191.Thatcher N., Hirsch F.R., Luft A.V., et al. Necitumumab plus gemcitabine and cisplatin versus gemcitabine and cisplatin alone as first-line therapy in patients with stage IV squamous non-small-cell lung cancer (SQUIRE): an open-label, randomised, controlled phase 3 trial. Lancet Oncol. 2015;16(7):763–774. doi: 10.1016/S1470-2045(15)00021-2. [DOI] [PubMed] [Google Scholar]
  • 192.Lynch T.J., Jr., Spigel D.R., Brahmer J., et al. Safety and effectiveness of bevacizumab-containing treatment for non-small-cell lung cancer: final results of the ARIES observational cohort study. J Thorac Oncol. 2014;9(9):1332–1339. doi: 10.1097/JTO.0000000000000257. [DOI] [PubMed] [Google Scholar]
  • 193.Chen Q., Fu Y.Y., Yue Q.N., et al. Distribution of PD-L1 expression and its relationship with clinicopathological variables: an audit from 1071 cases of surgically resected non-small cell lung cancer. Int J Clin Exp Pathol. 2019;12(3):774–786. [PMC free article] [PubMed] [Google Scholar]
  • 194.Reck M., Rodríguez-Abreu D., Robinson A.G., et al. Updated analysis of KEYNOTE-024: pembrolizumab versus platinum-based chemotherapy for advanced non-small-cell lung cancer with PD-L1 tumor proportion score of 50% or greater. J Clin Oncol. 2019;37(7):537–546. doi: 10.1200/JCO.18.00149. [DOI] [PubMed] [Google Scholar]
  • 195.Mok T.S.K., Wu Y.L., 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(10183):1819–1830. doi: 10.1016/S0140-6736(18)32409-7. [DOI] [PubMed] [Google Scholar]
  • 196.Paz-Ares L., Vicente D., Tafreshi A., et al. A randomized, placebo-controlled trial of pembrolizumab plus chemotherapy in patients with metastatic squamous NSCLC: protocol-specified final analysis of KEYNOTE-407. J Thorac Oncol. 2020;15(10):1657–1669. doi: 10.1016/j.jtho.2020.06.015. [DOI] [PubMed] [Google Scholar]
  • 197.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(2):220–233. doi: 10.1016/S1470-2045(21)00650-1. [DOI] [PubMed] [Google Scholar]
  • 198.Dearden S., Stevens J., Wu Y.L., Blowers D. Mutation incidence and coincidence in non small-cell lung cancer: meta-analyses by ethnicity and histology (mutMap) Ann Oncol. 2013;24(9):2371–2376. doi: 10.1093/annonc/mdt205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Lau S.C.M., Pan Y., Velcheti V., Wong K.K. Squamous cell lung cancer: current landscape and future therapeutic options. Cancer Cell. 2022;40(11):1279–1293. doi: 10.1016/j.ccell.2022.09.018. [DOI] [PubMed] [Google Scholar]
  • 200.Chen Y., Tang W.Y., Tong X., Ji H. Pathological transition as the arising mechanism for drug resistance in lung cancer. Cancer Commun. 2019;39(1):1–13. doi: 10.1186/s40880-019-0402-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Hassin O., Oren M. Drugging p53 in cancer: one protein, many targets. Nat Rev Drug Discov. 2023;22(2):127–144. doi: 10.1038/s41573-022-00571-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Lin J., Jin J., Shen Y., et al. Emerging protein degradation strategies: expanding the scope to extracellular and membrane proteins. Theranostics. 2021;11(17):8337–8349. doi: 10.7150/thno.62686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Redman M.W., Papadimitrakopoulou V.A., Minichiello K., et al. Biomarker-driven therapies for previously treated squamous non-small-cell lung cancer (Lung-MAP SWOG S1400): a biomarker-driven master protocol. Lancet Oncol. 2020;21(12):1589–1601. doi: 10.1016/S1470-2045(20)30475-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Eschweiler S., Ramírez-Suástegui C., Li Y., et al. Intermittent PI3Kδ inhibition sustains anti-tumour immunity and curbs irAEs. Nature. 2022;605(7911):741–746. doi: 10.1038/s41586-022-04685-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Ling Y., Liu J., Qian J., et al. Recent advances in multi-target drugs targeting protein kinases and histone deacetylases in cancer therapy. Curr Med Chem. 2020;27(42):7264–7288. doi: 10.2174/0929867327666200102115720. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Multimedia component 1
mmc1.docx (51.3KB, docx)

Articles from Genes & Diseases are provided here courtesy of Chongqing Medical University

RESOURCES