Hostname: page-component-745bb68f8f-mzp66 Total loading time: 0 Render date: 2025-02-06T08:57:08.192Z Has data issue: false hasContentIssue false

Cancer mutation profiles predict ICIs efficacy in patients with non-small cell lung cancer

Published online by Cambridge University Press:  04 April 2022

Liping Zhu
Affiliation:
Department of Medical Oncology, Shouguang Hospital of Traditional Chinese Medicine, Shouguang, Shandong Province, People's Republic of China
Dafu Ye
Affiliation:
Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
Tianyu Lei
Affiliation:
Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
Jie Wu
Affiliation:
Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
Wei Wang*
Affiliation:
Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
Bin Xu*
Affiliation:
Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China
*
Author for correspondence: Bin Xu, E-mail: xubin_oncology@whu.edu.cn, Wei Wang, wangwei1018@whu.edu.cn
Author for correspondence: Bin Xu, E-mail: xubin_oncology@whu.edu.cn, Wei Wang, wangwei1018@whu.edu.cn
Rights & Permissions [Opens in a new window]

Abstract

Although immune checkpoint inhibitors (ICIs) have produced remarkable responses in non-small cell lung cancer (NSCLC) patients, receivers still have a relatively low response rate. Initial response assessment by conventional imaging and evaluation criteria is often unable to identify whether patients can achieve durable clinical benefit from ICIs. Overall, there are sparse effective biomarkers identified to screen NSCLC patients responding to this therapy. A lot of studies have reported that patients with specific gene mutations may benefit from or resist to immunotherapy. However, the single gene mutation may be not effective enough to predict the benefit from immunotherapy for patients. With the advancement in sequencing technology, further studies indicate that many mutations often co-occur and suggest a drastic transformation of tumour microenvironment phenotype. Moreover, co-mutation events have been reported to synergise to activate or suppress signalling pathways of anti-tumour immune response, which also indicates a potential target for combining intervention. Thus, the different mutation profile (especially co-mutation) of patients may be an important concern for predicting or promoting the efficacy of ICIs. However, there is a lack of comprehensive knowledge of this field until now. Therefore, in this study, we reviewed and elaborated the value of cancer mutation profile in predicting the efficacy of immunotherapy and analysed the underlying mechanisms, to provide an alternative way for screening dominant groups, and thereby, optimising individualised therapy for NSCLC patients.

Type
Review
Copyright
Copyright © The Author(s), 2022. Published by Cambridge University Press

Introduction

Cancer is a disease characterised by abnormal proliferating cells caused initially by genetic mutations including non-small cell lung cancer (NSCLC) and other cancers. The causes of genetic mutations can be roughly divided into two categories: acquired mutations, including TP53, PTEN and other somatic mutations induced by ageing, poor lifestyles, environmental factors, etc., and inherited mutations, such as mutations in the BRAC1/BRAC2. In a retrospective study, researchers showed that 13.3% of cancer patients had inheritable cancer-related genetic mutations, including six most common types: BRCA1/BRCA2 mutation, MUTYH mutation, CHEK2 mutation, ataxia-telangiectasia mutated (ATM) mutation, mismatch repair (MMR) gene mutations including MLH1, MSH6, MSH2, MSH3, PMS2 (Ref. Reference Samadder1). In addition, most of the somatic mutations are passenger mutations but they still occupy the predominate gene mutations leading to the initiation of a tumour. Driver mutations are recognised to be the origin of malignances, such as encoding oncogenes (MYC, anaplastic lymphoma kinase (ALK), V-Ki-ras2 Kirsten rat sarcoma viral oncogene (KRAS), mesenchymal-to-epithelial transition (MET), epidermal growth factor receptor (EGFR), nuclear factor erythroid 2-like factor 2/Kelch-like ECH-associated protein 1 (NRF2/KEAP1), and BRAF) and tumour suppressors (TP53, PTEN, serine/threonine kinase 11/liver kinase B1 (STK11/LKB1), ATM) (Ref. Reference Martinez-Jimenez2). Genetic mutations or alterations affect a series of biological behaviours of tumour including tumour occurrence, development, invasion, metastasis, and so on (Refs Reference Martinez-Jimenez2, Reference Thomas3). And they remain widely prevalent in NSCLC. Thus, driver gene mutations become targets of therapy and biomarkers of predicting the efficacy of therapy, including ICIs, especially in NSCLC.

In the tumour microenvironment (TME), there're various genes exerting immunomodulation function via activating their downstream signalling pathways (Refs Reference Boussiotis4, Reference Lastwika5, Reference Zhang6, Reference Roelands7). The immune response involves tumour antigen exposure, antigen presentation, main histocompatibility complex (MHC) expression, the expression of cell surface receptors, the secretion of cytokines and chemokines, and the endocytosis and degradation of immune checkpoints (Refs Reference Wu8, Reference Chen and Mellman9, Reference Chen and Mellman10). At present, diverse approaches have been targeted to these processes to develop treatments for tumours, such as adoptive cell therapy (ACT), ICIs, cancer vaccines, and oncolytic viruses (Refs Reference Zhao11, Reference Kelly12). Nonetheless, the efficacy of current approaches remains limited. Previous studies suggested that co-mutations were effective biomarkers of immunotherapy (Refs Reference Wang13, Reference Marinelli14) and patients with 2 or more compound mutations were relevant to the significant benefit of ICIs (Ref. Reference Pan15). A genomic mutation signature encompassing eight genes (TP53, KRAS, STK11, EGFR, PTPRD, KMT2C, SMAD4, and HGF) was developed to predict the efficacy of ICIs in non-squamous NSCLC (Ref. Reference Bai16). Not only that, Sun et al. found that MGA mutation, a tumour suppressor gene was related to a response of ICIs (Ref. Reference Sun17). Our analysis from The Cancer Genome Atlas (TCGA) dataset also identified that the co-mutations and single mutations of KEAP1, KRAS, and STK11 indicated TME alteration (Ref. Reference Wu18). In molecular mechanism, the downstream pathways of these genes, including phosphatidylinositol 3 kinase (PI3 K)/AKT, MEK/extracellular regulated protein kinases (ERK)/mitogen-activated protein kinases (MAPK), MAPKs, signal transducer and activator of transcription (STATs), and nuclear factor-kappa B (NF-κB) etc., are closely correlated with an immune response (Refs Reference Zhang6, Reference Kumagai, Koyama and Nishikawa19). With the advances of sequencing technology, there is an expanding spectrum of identified oncogenic driver mutations in NSCLC. Based on these studies, the relationship between co-mutations and tumour sensitivity to immunotherapy needs to be further explored. Therefore, this review focused on the mono- and co-occurring genomic alterations as novel biomarkers of immunotherapy and sought to shed light on the underlying mechanism.

Current situation of immunotherapy

Although targeted therapy has inspiring therapeutic benefits in lung cancer patients with certain oncogenic mutations, the 5-year survival rate is still less than 20% due to the inevitable drug resistance and intra-driver heterogeneity (Refs Reference Siegel, Miller and Jemal20, Reference Skoulidis and Heymach21). Immunotherapy to revolutionise the cancer therapy, however, has overwhelmingly appeared and been widely utilised in lung cancer patients by inducing or reactivating antitumour immune response. For example, cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) is a coinhibitory molecule expressed on T lymphocytes which, upon a stronger affinity for CD80 and CD86 than for CD28, contributes to T cell anergy (Refs Reference Facchinetti22, Reference Rudd, Taylor and Schneider23). Programmed cell death ligand 1 (PD-L1) primarily exists in tumour cells and interacts with its receptor, programmed cell death 1 (PD-1), expressed notably on T lymphocytes. Once coupled with its receptor, PD-L1 inhibits CD8 T cell cytotoxic immune response to suppress the antitumour immune response (Refs Reference Barber24, Reference Xia, Liu and Wang25). Of note, a few of ICIs have been U.S. Food and Drug Administration (FDA)-approved through clinical trials, such as anti–CTLA-4 agent ipilimumab, tremelimumab and anti–PD-1/PD-L1 antibodies nivolumab, pembrolizumab, atezolizumab, durvalumab and avelumab (Ref. Reference Xia, Liu and Wang25). They have provided more personalised and accurate options for lung cancer patients, though the fact that the response rates of these ICIs only range from 14 to 20% in unselected patients and some even suffer severe immune-relate adverse events(irAEs) exists (Refs Reference Xia, Liu and Wang25, Reference Brahmer26, Reference Fehrenbacher27). Thus, highly specific, and sensitive predictive markers for ICIs are urgently needed to be found.

Currently, microsatellite instability (MSI) status and PD-L1 expression on tumour cells by immunohistochemical (IHC) stain are approved biomarkers to predict ICIs efficacy. Numerous studies have demonstrated that MSI is caused by the defect of the DNA MMR gene, which is closely related to the occurrence of tumours. MSI has been proved to improve immune-related objective response rate and immune-related progression-free survival rate in different types of solid tumours (Refs Reference Le28, Reference Le29), especially in colorectal cancers (Refs Reference Moreira30, Reference Sargent31). Thus, MSI is widely regarded as an important and effective marker for predicting the response of ICIs efficacy (Ref. Reference Le28). However, the researches of MSI in lung cancer are rare and the ratio of dMMR/MSI-H in lung cancers is very low and thus hasn't been widely used in lung cancer patients.

Testing for PD-L1 expression on tumour cells by IHC has a huge challenge because some PD-L1-negative patients exhibit a dramatic response to ICIs while a part of PD-L1-positive patients have no or low response (Ref. Reference Champiat32). In the KEYNOTE-024 trial (Ref. Reference Reck33), pembrolizumab monotherapy as the first-line therapy significantly improved progression-free survival (PFS), objective response rate (ORR) and overall survival (OS) in patients with tumours with PD-L1 ≥ 50%, while atezolizumab as the second-line therapy in metastatic patients, showed an improved survival versus docetaxel in all subgroups with different PD-L1 IHC assays and its benefit was independent of PD-L1 expression compared with pembrolizumab (Ref. Reference Rittmeyer34). Meanwhile, different detection platforms and assay conditions in distinct immunotherapy agents (Ref. Reference Osmani35), the inconsistent cut-points for PD-L1 expression and the heterogeneity of PD-L1 expression in different tumours (Refs Reference Bodor, Boumber and Borghaei36, Reference Zhou37) have indicated that it is a profound issue to identify more powerful predictive markers for ICIs response in NSCLC patients.

Subsequently, TMB as the potential predictive biomarkers for ICIs response, in view of the occurrence of neoantigen presented to reactivate immune responses, needs to be explored more. Previous studies have demonstrated the compelling evidence for TMB to predict the response rates of ICIs in various tumours including NSCLC across whole-exome sequencing (WES) or next-generation sequencing (NGS) to assess mutational burden through quantifying the number of non-synonymous mutations (Refs Reference Yarchoan, Hopkins and Jaffee38, Reference Rizvi39). Of note, the CheckMate 227 trial (Ref. Reference Hellmann40) elaborated the significant value of a high tumour mutational burden, independent of PD-L1 expression level in predicting response to the combination of ipilimumab with nivolumab. Nonetheless, a combination of a low TMB (<10 mutations per Mb) with a PD-L1 TPS <1% was regarded as exclusion criteria to exclude patients who were not benefiting from ICIs in CheckMate 227 trial (Ref. Reference Hellmann40). Likewise, Hellmann et al. presented compelling evidence that TMB is a potent biomarker with improved objective response, durable benefit, and progression-free survival, even OS in lung cancer patients (including NSCLC and SCLC) with combination immunotherapy though WES (Refs Reference Hellmann41, Reference Hellmann42, Reference Assoun43). Furthermore, TMB as a predictive marker exists other limitations such as the discrepancy between blood-based TMB and tissue-based TMB (Refs Reference Gandara44, Reference Cabel45, Reference Guibert46), the difference of testing platforms and the definite define of ‘higher’ TMB within various tumours (Refs Reference Bodor, Boumber and Borghaei36, Reference Camidge, Doebele and Kerr47). Although TMB-H (≥10 mut/Mb) was FDA-approved to screen progressed advanced NSCLC patients after receiving standard-of-care to use pembrolizumab based on the Keynote 158 study, patients of TMB-H did not show significantly improved PFS and OS.

With the knowledge of various predictive biomarkers of ICIs response, due to the wide genomic testing, many clinical trials found the association between genomic mutation and ICIs. In the following text, we will introduce the common mutations in NSCLC and demonstrate the value of their mutation profiles (especially co-mutations) as the independent predictors of immunotherapy and the potential and underlying mechanisms.

The major types of gene mutations in tumour and their downstream signals

Single mutations modulate the different signalling, including RAS/MEK/ERK/MAPK, PI3 K/AKT, STK11/adenosine monophosphate-activated protein kinase (AMPK)/mammalian target-of-rapamycin (mTOR), JAK/STAT3, phospholipase C gamma (PLCγ)/phosphatidylinositol-4,5-biphosphate (PIP2), and NF-κB etc. (Ref. Reference Rotow and Bivona48). These pathways participate in the signal transduction, cell circle, proliferation, apoptosis, and immune response (Refs Reference Baluapuri, Wolf and Eilers49, Reference Cuadrado50, Reference Kotler51, Reference Shaw and Engelman52, Reference da Cunha Santos, Shepherd and Tsao53, Reference Guertin and Sabatini54). Co-mutations may synergise to activate or suppress signalling pathways (Fig. 1). Therefore, we briefly summarised several signals of the driver gene in the following.

Fig. 1. The downstream signalling pathway of Tumour-related gene mutation. Bold fonts indicated that the protein existed corresponding gene mutation. Green represented suppressor gene while orange represented proto-oncogene.

TP53

TP53, the tumour suppressor genes, encoding the p53 transcription factor, is related to regulating various cancer-associated pathways including cell cycle arrest, DNA damage repair, metabolism, apoptosis, angiogenesis, and metastasis, and even inflammation and the immune response (Refs Reference Kotler51, Reference Kastenhuber and Lowe55). TP53 was commonly mutated in 46% lung adenocarcinoma (LUAD) patients according to TCGA data. Consistent with the role of p53 in tumour suppression, p53 protein is in low levels because of the regulators like MDM2. MDM2 is a p53 ubiquitin ligase to promote the degradation of P53 in normal cells while stimulus-dependent activities such as DNA damage facilitate p53 phosphorylation to activate p53's function through blocking MDM2-mediated degradation (Ref. Reference Kastenhuber and Lowe55). However, the study has demonstrated that the p53 gene is transcriptionally induced by IFN-α/β through IFN-activated transcription factor interferon-stimulated gene factor 3 (ISGF3) activation, demonstrating that p53 gene is induced by a cytokine (Ref. Reference Takaoka56). Likewise, toll-like receptors (TLR) gene expression pattern could be changed by the presence of p53 in damage situation (Ref. Reference Shatz, Menendez and Resnick57), which shows the relationship between p53 mutation and immune response. TP53 mutations have been identified as controversial biomarkers for ICIs in NSCLC patients due to the co-occurring mutations and mutation subtypes (Ref. Reference Sun58), while MDM2/MDM4 amplifications have been proved to be correlated with hyperprogression with anti-PD-1 therapy (Ref. Reference Kato59).

KRAS

KRAS, as a member of the GTPase family, is one of the most frequently oncogenes aberrations in NSCLC, occurring in 0%~33% of adenocarcinomas (Ref. Reference Cancer Genome Atlas Research60). Despite the KRAS-MAPK as the downstream of EGFR signalling, KRAS activation still signals through the mitogen-activated protein kinase and PI3 K/AKT/mTOR cascade mediated by EGF, irrespective of EGFR state (Refs Reference Rotow and Bivona48, Reference Martin Martorell61, Reference Roman62). KRAS mutations in lung cancer have been regarded to be mutually exclusive driver mutations on behalf of a genetically heterogeneous subgroup due to the variety of KRAS-mutations and co-mutations (Ref. Reference Scheffler63). Strikingly, up to now, Sotorasib is the only FDA-approved tyrosine kinase inhibitors (TKIs) targeting KRASG12C, which is a milestone of KRAS-specific inhibitor (Ref. Reference Hong64). A previous meta-analysis suggested that NSCLC patients with KRAS mutations responded well to ICIs compared with KRAS-wild types (p = 0.001) (Ref. Reference Landre65). This may be attributed to that KRAS mutations are closely associated with smoking, so as to increase the expression of neoantigens and TMB levels (Ref. Reference Scheel66).

PI3 K/PTEN

PI3 K-AKT signalling is involved in various biological processes in many types of cancer, its overactivation contributes to abnormal cell cycle progression, epigenetic modification, adhesions and motility changes, inhibition of apoptosis and induction of angiogenesis (Refs Reference Yang, Jiang and Hou67, Reference Fruman68) by activating mutations in PIK3CA, loss of PTEN and so on. PIK3CA gene encodes a catalytic subunit, p110α, which catalyses PIP2 into phosphatidylinositol triphosphate (PIP3) to mediate the phosphorylation of Akt at thr308 by promoting PDK1 (phosphoinositide-dependent kinase 1) and Akt interaction, and thus activating downstream signalling (Ref. Reference Alzahrani69). While the tumour suppressor gene PTEN negatively regulates the PI3 K/AKT activation by encoding a dual protein/lipid phosphatase (Ref. Reference Vazquez and Devreotes70). PTEN dephosphorylates PIP3 to PIP2 to antagonises the function of PI3 K, and thus, the loss of PTEN also contributes to the activation of Akt (Refs Reference Alzahrani69, Reference Vazquez and Devreotes70). There were 7% LUAD patients harbouring PI3KCA mutation (Ref. Reference Cancer Genome Atlas Research60). There is a lack of data to find out the association between PI3KCA mutation and ICIs efficacy in NSCLC patients.

KEAP1/NRF2

NRF2 is encoded by NFE2L2, a member of a transcription factor family which can be translocated into the nucleus and regulate the transcription of target genes (Ref. Reference Ahmed71). KEAP1, recognised as the principle negative regulator of NRF2, is a cysteine-rich and redox-sensitive protein, binding to NRF2 and inducing the CUL3-mediated ubiquitination and proteasomal degradation (Ref. Reference Ahmed71). The KEAP1/NRF2 axis is pivotal in the defence mechanism against oxidative and electrophilic stress by eliciting antioxidant, detoxification, and anti-inflammatory proteins (Ref. Reference Cuadrado50). Thus, alterations in the KEAP1 and NRF2 genes mainly contribute to the constitutive activation of NRF2, which accounts for the activation of downstream target genes for cell proliferation and resistance to therapy (Ref. Reference Hammad72). The mutation of KEAP1/NRF2 was found in 17% LUAD patients (Ref. Reference Cancer Genome Atlas Research60), have a poor prognosis and may not benefit from anti-PD-L1 treatment (Ref. Reference Singh73). Meanwhile, this study demonstrated that KEAP1 mutation synergises with alterations in STK11 and KRAS to represent a more aggressive NSCLC subtype in promoting early-onset, initiation, and proliferation of tumour (Ref. Reference Singh73). The underlying mechanism may be related to immune escape in the tumour immune microenvironment regulated by KEAP1 (Ref. Reference Best and Sutherland74), which promoted the cytokines secretion including IL-6 and IL-11 and reduced leucocyte infiltration (Ref. Reference Matsuoka75).

STK11/LKB1

STK11/LKB1 encodes a serine-threonine kinase, which is recognised as a tumour suppressor and a regulator in the metabolism of glucose and lipid in cells, cell growth and polarity through phosphorylation of AMPK and 12 AMPK-related kinases (Ref. Reference Shackelford and Shaw76). STK11 mutation occupied about 17% LUAD patients (Ref. Reference Cancer Genome Atlas Research60). The downstream pathway of STK11/AMPK is the mTOR pathway suppressed by AMPK phosphorylation of TSC2 and raptor and mTOR complex 1 (mTORC1) modulates the expression of cell growth regulators, such as cyclin D1, hypoxia-inducible factor 1α (HIF-1α), and C-Myc, all of which relate to tumorigenesis and immune response (Ref. Reference Guertin and Sabatini54). Other associations between STK11, AMPK and p53 have been identified, including directly or indirectly activating p53 through phosphorylation (Ref. Reference Shackelford and Shaw76). Nonetheless, STK11 mutations in NSCLC patients may not respond to ICIs therapy (Ref. Reference Rizvi39). Research has shown that STK11/LKB1 deficiency led to an immunosuppressive tumour microenvironment by increasing neutrophil recruitment and reducing T cell activity in the TME (Ref. Reference Koyama77).

EGFR

EGFR gene encodes a type I transmembrane growth factor receptor with the tyrosine kinase domain, mutant EGFR has been proven to contribute to increased proliferation, metastasis, angiogenesis, and decreased apoptosis by activating the PI3 K-PTEN-AKT and RAS/MEK/ERK/MAPK pathways (Ref. Reference da Cunha Santos, Shepherd and Tsao53). It's reported that EGFR mutation only occurred 14% in lung adenocarcinoma in TCGA data (Ref. Reference Cancer Genome Atlas Research60). Nonetheless, cancers with EGFR alterations generally exhibit the non-inflamed TME with reducing infiltrating CD8+ T cells and so on (Ref. Reference Kumagai, Koyama and Nishikawa19), via PI3 K/AKT pathways. Thus, the studies demonstrated that NSCLC patients with EGFR mutation had poor response to ICIs in NSCLC due to the uninflamed TME with weak immunogenicity, low T cell infiltration and low PD-L1 expression (Refs Reference Dong78, Reference Gainor79).

BRAF

BRAF as a serine/threonine-protein kinase, its mutations occur in 3–10% of lung adenocarcinoma, the most common mutation of which is the BRAFV600E mutation (Refs Reference Rotow and Bivona48, Reference Cancer Genome Atlas Research60), others including BRAFG469A/V and BRAFD594 G mutations. Constitutive BRAF activation induced by BRAFV600E mutation activates downstream MEK/ERK signalling pathway, thus promoting tumour proliferation and growth (Refs Reference Dankner80, Reference Negrao81). The combination of MEK and BRAF-V600-specific inhibitors further enhance treatment outcomes in BRAF-V600E-positive NSCLC patients (Ref. Reference Planchard82). However, there are still approximately 50% non-BRAF-V600E-mutated NSCLC patients who cannot receive specific BRAF-inhibitor (Ref. Reference Negrao81), indicating that it is an urgent need to explore the potency of immunotherapy application in these patients. The research showed that NSCLC patients harbouring BRAF mutations showed limited response to ICIs although these mutations were associated with high expression of PD-L1 and low/intermediate TMB level (Refs Reference Dudnik83, Reference Murciano-Goroff84).

MET

Activating mutation and genomic amplification were discovered in the MET gene, a novel and potential therapeutic target in NSCLC (Ref. Reference Trusolino, Bertotti and Comoglio85). Meanwhile, a mutation in MET was variable, including exon 14 skipping and amplification. Both mutations occupied less than 7% NSCLC patients (Refs Reference Cancer Genome Atlas Research60, Reference Awad86). MET as a member of proto-oncogenes, encodes a receptor tyrosine kinase binding of hepatocyte growth factor (HGF), inducing downstream RAS-ERK/MAPKs, PI3 K/Akt, STAT, and NF-κB cascades (Ref. Reference Trusolino, Bertotti and Comoglio85). MET mutations were modestly correlated with the efficacy of ICIs treatment in NSCLC patients in spite of high PD-L1 expression (Refs Reference Li, Wang and Wang87, Reference Sabari88). There is evidence that NSCLC patients harbouring MET exon 14 mutation exhibited remarkably low TMB levels, which is similar to BRAF mutant NSCLC and MET exon 14 mutation contributed to an immunosuppressive TME by inhibition of DCs and suppression of T-cell proliferation induced by neutrophils (Refs Reference Sabari88, Reference Papaccio89).

ALK

The echinoderm microtubule-associated protein-like 4 (EML4) and ALK fusion gene was initially found in NSCLC patients by Soda, resulting from an inversion in chromosome 2 (Ref. Reference Golding90). The inversion protein mediates constitution activation of ALK by ligand-independent dimerisation. The activation of RAS/MEK/ERK, JAK/STAT3, PLCγ and PI3 K/AKT pathways are vital signalling pathways contributing to cellular proliferation and growth, which can be modulated by ALK activity (Ref. Reference Shaw and Engelman52). However, in TCGA data, ALK fusion gene mutation was found only in 1% LUAD patients (Ref. Reference Cancer Genome Atlas Research60). NSCLC patients harbouring ALK rearrangements were associated with a low response to ICIs due to the low PD-L1 expression and low infiltration of CD8+ tumour infiltrating lymphocytes (Ref. Reference Gainor79).

ATM

ATM gene, initially mutated in the autosomal recessive disease ataxia–telangiectasia (AT), encodes a 370-kDa protein of the PI3 K superfamily and activates downstream protein targets in different cell circles when it senses double-strand breaks of cellular DNA damage caused by ionising radiation (IR) and other agents (Refs Reference Bakkenist and Kastan91, Reference Mitui92). Whereas, the activation of ATM is correlated with the protein serine–threonine phosphatase 5 (PP5) (Ref. Reference Ali93). Studies also have illustrated that ATM and XRCC4-like factor (XLF) have redundant functions in the process of V(D)J recombination in developing lymphocytes and ATM and XLF co-deficiencies contribute to a severe combined immunodeficiency (SCID) phenotype similar to that observed in the background of canonical-nonhomologous end-joining (C-NHEJ) deficiency (Ref. Reference Kumar, Alt and Oksenych94). Thus, ATM plays a pivotal role in various pathways including genomic instability, cancer susceptibility, as well as immunodeficiency and so on. However, there is a lack of research to study the role of ATM mutation in NSCLC and in NSCLC patients treating with ICIs.

Myc

Myc oncoproteins are regarded as transcription factors with three members in its family, C-Myc, N-Myc, and L-Myc (Ref. Reference Dang95), which is amplified in approximately 3.3% of LUAD patients. They are called ‘super-transcription factors’ due to latently modulating the transcription of more than 15% of the entire genome (Ref. Reference Dang96). Thus, Myc oncogenes regulate a number of biological functions, including cell proliferation, differentiation, survival, as well as immune surveillance through orchestrating downstream effectors, such as ribosome synthesis, protein translation, metabolism and cell cycle progression (Ref. Reference Chen, Liu and Qing97). The underlying mechanism of Myc oncoproteins to promote cell proliferation and tumourigenesis may not only alter target gene expression but also change the basic transcription mechanisms by modulating transcription elongation with cell cycle progression (Ref. Reference Baluapuri, Wolf and Eilers49). In addition, Myc has been discussed to regulate the association between tumour cells and immune cells by controlling the synthesis of relevant cytokines. Nonetheless, Myc mutation reprogrammed the tumour immune microenvironment by loss of T cells, NK cells and B cells mediated by CCL9 and IL-23 (Ref. Reference Kortlever98). Their relationship with ICIs efficacy is still unclear.

Although most of the single gene mutations, including TP53, KRAS, KEAP1, STK11, BRAF, MET and ALK, have been verified to be related to the efficacy of ICIs, not all mutations of them have equivalent effects due to the different mutation subtypes among these mutations. For example, TP53 missense has shown better clinical benefits from anti-PD-1/PD-L1 therapy compared with nonsense mutants. In addition, co-occurring mutation subtypes such as mutations in KRAS and other genes (STK11 or TP53) also have different influences on the ICIs efficacy in NSCLC. Next, we predominantly reviewed the research of co-mutations in NSCLC patients receiving ICIs to explore the independent predictors of immunotherapy and the potential and underlying mechanisms.

The co-mutation subtypes related to ICIs

The Underlying mechanism of co-mutation

The underlying mechanisms of genetic mutations including oncogenic mutation and suppressor genetic mutation are very complicated. Currently, a study (Ref. Reference Sanchez-Vega99) analysed ten typical and classical pathways with commonly altered genes to explore the patterns of reoccurrence, co-mutations, and mutual exclusivity. The results were that co-mutation events mediated synergistic activation of each pathway such as LKB1 and KEAP1/NRF2 pathways synergically driving glutamine-dependent metabolic reprogramming (Ref. Reference Galan-Cobo100) and explicitly showed that some pathways were markedly mutually exclusive such as RTK-RAS pathway while others often had co-occurring mutations per tumour including PI3 K and NRF2 pathways (Ref. Reference Nissan101). Moreover, resistance to therapy was also reflected by patterns of co-mutation in many tumours when targeting one of the alterations (Ref. Reference Nissan101). For example, TP53 co-mutation in Her2-mutant LUAD cause resistant to Afatinib (Refs Reference Yuan102, Reference Fang103). Major studies have identified that mutations are mutually exclusive due to pathway structure which may correlate with functional redundancy.

Genetic epistasis may account for that a driver mutation is unlikely to occur after an existing mutation with common or superfluous functional effects in the same molecular pathway due to non-additively to tumour fitness in the same pathway with multiple genes (Refs Reference Mina104, Reference van de Haar105). Furthermore, the latter mutation will not provide a further evolutionary advantage of tumours in the presence of the prior mutation because of the effect of synthetic lethality, as cells cannot survive with both alterations (Ref. Reference Etemadmoghadam106). Thus, it may be promising to exploit and identify the second-site targets including oncogenic and non-oncogenic mutations, which are aberrant in alliance with a tumour-specific mutation, based on synthetic lethality. For instance, poly (ADP-ribose) polymerase (PARP) inhibitors are used to treat BRCA1/2 mutated ovarian cancers (Refs Reference Brown107, Reference George, Kaye and Banerjee108) and together with MYC blockade to treat triple-negative breast (Ref. Reference Carey109).

Of note, Haar et al. (Ref. Reference van de Haar105) illustrated that mutually exclusive mutations also resulted from interactions with disease subtype and tumour mutation load through pan-cancer analysis. It demonstrated that mutations contributing to low mutation load in tumour subtype might be mutually exclusive. For example, oncogene EGFR with low tumour mutation load in LUAD of nonsmoking patients presented with mutual exclusivity (Ref. Reference Cancer Genome Atlas Research60). ‘Mutation load association’ (MLA) was calculated to estimate the higher mutation frequencies in cancers with low or high mutation loads. In addition, due to different cancer types, gene mutation status was significantly distinct in different pathways. Thus, the specific MLA of each gene correlated with cancer type. Researchers speculated that gene with low MLA was more likely to be mutual exclusive by correcting for tumour type. Moreover, driver mutation might tend to have a low MLA while passenger mutation might positively correlate with higher MLA. In conclusion, the results demonstrated that complicated interactions between mutation loads, gene mutation frequencies, gene mutation state, gene mutation types and tumour subtypes contributed to a complex system in which both direct gene-gene interactions (epistasis) and indirect causal paths, which might drive patterns of mutual exclusivity or co-mutation between mutations (Ref. Reference van de Haar105).

The co-mutation subtypes and predictive role of ICIs

Generally, EGFR mutation is mutually exclusive. However, Yang et al. found patients with EGFR-MAPK co-mutations had increasing levels of both TMB and PD-L1 protein expression through analysing data from TCGA and The Cancer Proteome Atlas (TCPA), which might identify a subgroup of NSCLC patients benefiting from ICIs with longer disease-free survival (DFS) (median DFS: EGFR-Mut: WT: Co-Mut = 1.87: 3.77: 7.82 (months), p-value = 0.03) (Ref. Reference Yang110). Nonetheless, due to the advances and benefits of TKIs, scarce studies explore the efficacy of ICIs in NSCLC patients with EGFR mutation or co-mutation. The KRAS-mutation is the opposite, and there has been no effective drugs targeting KRAS mutation sites in the past except Sotorasib, which makes it difficult to treat. Thus, KRAS-mutation was reported to have a causal relationship with immunoresistance in the lung cancer microenvironment with coexisting alterations and correlated with poor prognosis. For co-mutation, Kortlever et al. initially identified that Myc and KRAS co-mutation elicit immunosuppressive TME through exclusion T, B, and NK cells in the mice model (Ref. Reference Kortlever98). Furthermore, studies also defined KRAS-STK11/LKB1 (KL) or KRAS-TP53 (KP) co-mutations as distinct subgroups of KRAS-mutant LUAD patients. KL tumours were recognised to be resistant to PD-1/PD-L1 inhibitors while KP LUAD were relatively responsive to PD-1/PD-L1 inhibitors (mOS: KP: K-only: KL = 6.4: 16.1: 16.0 (months), p-value = 0.0045) (Ref. Reference Skoulidis111). What is more, studies demonstrated that KP lung cancers associated with epithelial–mesenchymal transition (EMT) were more sensitive to PD-1/PD-L1 axis inhibitors than epithelial KP tumours (Ref. Reference Peng112). Additionally, co-mutation of KEAP1/NFE2L2 and KRAS, as an independent prognostic factor, demonstrated worse OS in NSCLC patients with ICIs (mOS: KRAS-KEAP1/NFE2L2: K-only = 10: 24 (months), p-value < 0.001), irrespective of the level of TMB (Ref. Reference Arbour113). However, not all KRAS mutations were equal in predicting the efficacy of ICIs in LUAD patients. For example, KRASG12C-TP53 co-mutation identified the positive responders to PD-1 inhibitor pembrolizumab in PD-L1 high (≥50%) LUAD patients ICIs (mOS: KRASG12C-TP53mut: KRASother-TP53wt = NE: 22.2 (months), HR for mOS: 0.17 (95% CI 0.04–0.76), p-value = 0.02) while KRASG12D/TP53 co-mutation created an immunosuppressive microenvironment and might be a negative predictive biomarker for anti-PD-1/PD-L1 ICIs in LUAD (Refs Reference Frost114, Reference Gao115). Additionally, KEAP1-PTEN co-mutation also exhibited an immunosuppressive microenvironment in the LUAD of mice model, along with the high expression of PD-L1, which might be sensitive to ICIs (Ref. Reference Best116). A sustained PI3 K signalling in combination with the activated KEAP1 pathway might facilitate lung tumourigenesis (Ref. Reference Best116). KEAP1 and STK11 co-mutation was reported to increase the risk of death in NSCLC patients with ICIs, such as in the tNGS cohort (HR of Co-Mut vs. WT for OS: 1.73 (95% CI 1.17–2.57)), thus as a predictor to recognise population of NSCLC patients unresponsive to ICIs (Ref. Reference Marinelli14).

Likewise, TP53 mutations are also varied and distinct. For example, TP53 missense and nonsense mutations were correlated with elevated neoantigen and TMB levels, and lead to DNA damage repair deficiency (Ref. Reference Assoun43). However, TP53 missense but not nonsense mutants were associated with better clinical benefits from anti-PD-1/PD-L1 therapy (Ref. Reference Assoun43). Thus, only such TP53 subgroups are expected to respond well to ICIs. A current study found that in lung cancer patients with DSPP and TP53 co-mutation subtype had a better PFS of immunotherapy with significantly infiltrated CD8 + T cell and decreased M2 macrophage level compared with wild-type (mPFS: co-mutation: WT = 9.3:4.9 (months), p-value = 0.008) (Ref. Reference Jiang117). Not only that, lung cancer patients harbouring TP53/Histone-lysine N-methyltransferase 2C (KMT2C) co-mutation also had significant benefit from ICIs (HR of Co-Mut vs WT for DFS: 0.48 (95% CI 0.24–0.94)) (Ref. Reference Shi118). TP53 and ATM co-mutation was correlated with better OS compared with a single mutation or no mutation in NSCLC patients treated with ICIs (mOS: TP53-ATM co-mutation: TP53 mutation alone: ATM mutation alone: no mutation = not reached: 11.0: 16.0: 14.0 (months), p-value = 0.24) (Ref. Reference Chen119). In patients with TP53 and ATM co-mutation, other mutations such as EGFR or ALK occurred in parallel without concurrent or exclusive patterns (Ref. Reference Chen119).

Notably, TP53 and ATM are also members of the DNA damage response (DDR) pathway, other co-mutations in which were also demonstrated to associate with the efficacy of ICIs in NSCLC patients, such as homologous recombination repair and mismatch repair (HRR-MMR) or HRR and base excision repair (HRR-BER) (Ref. Reference Wang13). Moreover, co-mutation between other signal pathways, such as NOTCH, and homologous repair genes were also reported to associated positively with the efficacy of ICIs in patients with advanced NSCLC (Ref. Reference Mazzotta120). The DDR systems contain eight pathways: MMR, BER, check point factors, Fanconi anaemia, HRR, nucleotide excision repair (NER), nonhomologous end-joining, and DNA translesion synthesis (Refs Reference Wang13, Reference Scarbrough121). DDR genes are responsible to regulate genetic instability and susceptibility due to its involvement in the damaged repertoire of DNA repair and DNA-damage signalling capabilities. The alteration in the DDR system will lead cells to fail to protect the genome against endogenous and exogenous damage (Ref. Reference Brown107). Therefore, many gene mutations in DDR pathways have been identified to relate with tumourigenesis, such as MMR (MLH1, MSH2, MSH6, as well as PMS2), BER (POLE), HRR (BRCA2), and ATM (Ref. Reference Wang13). Overall, DDR pathway alteration is related to tumour mutation burden and tumour-specific neoantigen load, which may be the explanation of its predictive value in ICIs efficacy in cancer. Moreover, the gene mutation disrupting the DDR pathway will present as high mutation loads and indicate benefit from ICIs, such as KMT2C mutation (Refs Reference Chang122, Reference Zhang and Huang123).

However, not all co-mutations have been defined to be a biomarker of ICIs in NSCLC patients. For instance, the correlation between MET exon 14 skipping and TP53 co-mutation and immunotherapy hasn't been identified although they are associated with increased PD-L1 expression (Refs Reference Wong124, Reference Albitar125). TP53 and EGFR co-mutation is also lack of evidence for predicting the response of ICIs (Ref. Reference Labbe126) although the co-mutation is closely associated with poor prognosis in LUAD (Ref. Reference Zheng127). The study demonstrated that EGFR and co-mutational tumour suppressor genes (TP53, KEAP1, STK11, etc.) had increased TMB, as an independent subtype of LUAD (Ref. Reference Zhao128). To date, the sparse study evaluates the association between EGFR co-mutation with the efficacy of ICIs. Besides, the value of KRAS mutation patients harbouring with CDKN2A/B or PIK3CA mutations in predicting the ICIs response is also still uncertain (Refs Reference Gu, Xu and Chang129, Reference Wang130). In total, the relationship between co-mutation and efficacy of immunotherapy may rely on mutation subtypes, mutation sites, a function of mutant genes, and their association with TMB and immune response, which needs a more comprehensive evaluation.

Potential Mechanisms of predicting ICIs by co-mutation

Although NSCLC patients with driver gene mutations are usually regarded to have a low response to ICIs (Refs Reference Gainor79, Reference Lee131), several driver genes have participated in the signalling pathway related to immune response and the regulation of tumour immune microenvironment (Refs Reference Koyama77, Reference Cho132, Reference Chen133, Reference Shamalov134, Reference Topper135, Reference Parkes136) (Figs 2 and 3). For instance, evidence from several research studies demonstrated that tumour suppressor proteins participate in modulating immune response and regulating the expression of immune checkpoints proteins. Thiem et al. found that P53 played a pivotal role in boosting IFN-γ-induced PD-L1 expression by inducing JAK2 expression (Ref. Reference Thiem137). IFN-γ produced by CD8+ cells was also reported to elicit the expression of the immune checkpoints in contrast to attracting cytotoxic immune cells, including PD-L1 on tumour cells and indoleamine-2,3-dioxygenase (IDO) on dendritic cells or macrophages, which promoted tumour evading and created the immunosuppressive TME. Besides that accumulated IFN-γ was recognised as the best incentive for PD-L1 expression, IL-17, as a proinflammatory cytokine secreted by Th17 cells (Ref. Reference Lei138), has also been reported to regulate the expression of PD-L1 or modulate the level of CD38 to facilitate immunotherapy resistance in KP lung cancer (Ref. Reference Peng112).

Fig. 2. The immune microenvironment of tumour. The tumour immune microenvironment changed with the onset and evolution of tumour. Tumour antigen was presented to CD8+ T cells directly or presented to CD4+ T cells indirectly by APC (antigen-presenting cell) to elicit anti-tumour response. M1 macrophage cells also exerted anti-tumour immune response. Additionally, tumour cells also secreted chemokines such as CXCL10 to recruit lymphocytes. Tumour-associated macrophage, Tregs and MDSC play a role in suppressing anti-tumour immune response through secreting TGF-β, IL-10, and expressing IDO-1 and IDO-2. Neutrophils secreted neutrophil elastase (NE) to promote tumour proliferation. Meanwhile, cellular matrix mediated by TGF-β in tumour blocked the infiltration of lymphocytes. Cancer-associated fibroblasts (CAFs) released stromal cell derived factors, angiogenic factors to promote tumour cell growth and tumour blood vessel regeneration.

Fig. 3. The pathway of gene mutation to influence the tumour-associated immune response. The diverse signalling pathways and transcriptional factors promoted the expression of PD-L1 to assist tumour to evade immune response. Tumour cells evade anti-tumour effects of T cells in part by elevating PD-L1 mRNA expression at transcriptional level via activation of different upstream signalling pathways.

What's more, cell-intrinsic oncogenic signalling, such as EGFR, was reported to facilitate PD-L1 expression in cancer cells via PI3 K-AKT-mTOR pathway (Ref. Reference Lastwika5). In addition, studies showed that EGFR-TKI resistant NSCLC may respond to ICIs therapy, based on the expression of PD-L1 (Refs Reference Nogami139, Reference Peng140). Thus, resistant mechanisms in EGFR mutation of increasing PD-L1 expression and attenuating immune response were identified (Ref. Reference Peng140). MAPK, PI3 K and NF-κB pathways participated in resistance mechanism-induced PD-L1 overexpression, such as HGF, c-MET amplification and EGFR-T790 M resistance mechanisms (Ref. Reference Peng140). EGFR co-occurring alterations with interferon regulatory factor 1 (IRF1) was reported to downregulate CXC-chemokine ligand 10 (CXCL10) and thus decrease effector CD8+ T cells infiltration (Refs Reference Kumagai, Koyama and Nishikawa19, Reference Matsuzaki141). The activation of MYC also indirectly increased the expression of PD-L1 in tumour cells through the influx of Chemokine (C-C motif) ligand 9 (CCL9)-dependent macrophage while Myc and KRAS co-mutation in LUAD haven't shown the similar mechanism to exert the immunosuppressive tumour environment (Ref. Reference Kortlever98). Meanwhile, the studies identified that RAS signalling upregulated PD-L1 expression in tumour cells through inhibition of the AU-rich element-binding protein tristetraprolin (TTP) via MEK signalling to increase the stability of PD-L1 mRNA (Refs Reference Chen133, Reference Coelho142).

DDR gene mutations, leading to unpaired DNA lesions and S-Phase-Specific DNA Damage, may enhance the accumulation of cytosolic DNA, which activates the stimulator of interferon genes (STING)-mediated pathway and then induces the expression of type I IFN, indicating neoantigen-independent pathways to activate innate antitumour immunity and boost the immune recognition and response. What's more, DDR-deficient molecular subtype may promote the expression of PD-L1 both in tumour cells and infiltrating lymphocytes (Refs Reference Barber143, Reference Hartlova144). The HR mutation in DDR gene mutations contributed to increased TMB and neoantigen exposure, enhancing the immune response against tumours while NOTCH was associated with the development, maintenance, and activation of T cells, in which co-mutation will represent the distinct subtype sensitive to ICIs in NSCLC (Ref. Reference Mazzotta120). Conversely, MET amplification decreased STING expression through inducing phosphorylation of UPF1 and attenuated the IFN response. Thus, the tumour infiltrating CD8+ T and NK cells were reduced (Ref. Reference Zhang145).

STK11 mutation was initially shown to upregulate the expression of KRAS genes and enhance the KRAS-induced signalling and gene expression responses for driving tumourigenesis. And in parallel it was correlated with reduced T cells in tumours likely through reducing the activity of NF-κB pathways, a pivotal immune regulatory pathway to mediate inhibition of immune surveillance (Ref. Reference Schabath146). Additionally, co-mutation STK11 and KRAS in a mouse model contributed to the recruitment of neutrophils with T cell suppressive effects and increasing the levels of T cell exhaustion markers (PD-1, CTLA-4, and T-cell immunoglobulin and mucin-domain containing-3 (TIM3)) as well as tumour-promoting cytokines by interleukin-6 (IL-6) (Ref. Reference Koyama77). In addition, STK11/LKB1 loss contributed to significant silencing of STING expression which may be insensitivity to cytoplasmic double-strand DNA (dsDNA) (Ref. Reference Kitajima147). Thus, co-occurring KRAS and STK11/LKB1 alterations reduced the level of innate immune signalling including inhibition of cytotoxic type 1 IFNs and chemokines CXCL10 which promoted T-cell recruitment by silencing STING and thus, promoted tumour immune escape (Refs Reference Barber143, Reference Kitajima147). Importantly, they correlated with low expression of PD-L1 whether in a mouse model or in patient tumours (Ref. Reference Koyama77). Thus, STK11 was associated with primary immunotherapy resistance. Likewise, in a cohort of NSCLC patients harbouring STK11/KEAP1 alterations, patients manifested as lower T cell receptor (TCR) richness and diversity and lower expression of Th1 and immunomodulatory genes, but had higher wound healing subsets compared with wild type, which was regarded as predictors of poor survival.

TP53 mutation causing loss of TP53 function, co-occurring with KRAS or not, was closely correlated with increased TMB and significantly enhance the tumour genomic instability to generate tumour neo-antigens, thereby eliciting tumour immunogenicity with increasing recruitment of cytotoxic T lymphocytes into TME, thus improving the efficacy of ICIs (Refs Reference Shamalov134, Reference Schumacher and Schreiber148). Neutrophils and dendritic cells were also closely correlated with TP53 mutation status (Ref. Reference Zhao149). Moreover, studies also demonstrated that altered unfold TP53 proteins caused by destabilising TP53 may also become tumour-specific antigens presented by class I MHC molecules, which could augment T cell reactivity (Ref. Reference Shamalov134). In contrast, Cha et al. has shown that p53 aberrant mutation assessed by IHC was closely correlated with PD-L1 expression in tumour cells (Ref. Reference Cha150). Likewise, p53 was proved to modulate PD-L1 expression by regulating the transcription of miR-34, a specific microRNA binding to PD-L1 in p53R172HΔg/ + K-rasLA1/+ syngeneic mouse model with lung adenocarcinoma. However, the inert relationship among them needs more evidence from clinical specimens to verify. Strikingly, TP53 and KRAS co-mutation presented with increased expression of PD-L1, and decreased expression of other non-PD-L1 immune inhibitory checkpoints, including Lymphocyte Activating 3 (LAG3) and V-Set Domain Containing T Cell Activation Inhibitor 1 (VTCN1), indicating a potential population benefiting from ICIs (Ref. Reference Cortez151).

In conclusion, the aberrant co-mutations of genes synergistically contribute to the immune function disorder to promote tumour immune evasion by influencing the immune checkpoint expression, the level of cytokines and inflammatory factors. The underlying complex mechanism may account for why patients with single gene mutation have no response to ICIs, but co-mutations have a surprising effect on predicting the response of ICIs.

The Relationship between co-mutations and current biomarkers of immunotherapy

A large body of studies has revealed that both single mutation and co-mutation affect the current biomarkers of immunotherapy, such as PD-L1, TMB and so on (Refs Reference Skoulidis and Heymach21, Reference Gao115, Reference Shi118). For example, EGFR mutations are correlated with a lower TMB (Ref. Reference Singal152) and lack of tumour infiltrating lymphocytes (TIL) (especially PD-L1 + /CD8 + TIL) that contributes an uninflamed TME and weak immunogenicity (Ref. Reference Dong78), suggesting an inferior response to ICIs in NSCLC patients (Refs Reference Dong78, Reference Gainor79). Interestingly, the aberrant mutations in EGFR can also drive PD-L1 expression, which indicates potential benefits from immunotherapy (Ref. Reference Gettinger and Politi153). The discrepancy between the above studies seems unable to determine whether patients with EGFR mutation can benefit from immunotherapy, so we expect more clinical/pre-clinical studies to demonstrate the response of ICIs in these patients. Meanwhile, we also lack the data of the PD-L1 expression, TMB, and TIL in different EGFR co-mutation subtypes.

Dong et al. found that TP53 and KRAS co-mutation had a possible synergistic effect on boosting PD-L1 expression (Ref. Reference Dong154). They also correlated with increased TMB level, accelerated T cell infiltration, as well as enhanced tumour immunogenicity (Ref. Reference Dong154). Likely, higher TMB and copy number alteration (CNA) are presented by co-occurring TP53- or STK11-KRAS co-mutation than KRAS mutation alone. However, as mentioned above, the biological phenotype may differ among different KRAS mutation sites. However, KRASG12D/TP53 co-mutation showed decreased TMB, PD-L1 expression, and immune cells infiltration, consisting of activated CD4 memory T cells, T helper cells, NK cells and M1 macrophages (Ref. Reference Gao115). KMT2C was associated with a higher level of TMB and higher PD-L1 expression compared with the wild type due to the genetic instability (Ref. Reference Shi118). In this regard, Zhao et al. demonstrated that truncating TP53 mutations were associated with poor survival in NSCLC patients in ICIs therapy with low TMB (Ref. Reference Zhao149), indicating that the TP53 mutation status may associate with different prognostic outcomes in NSCLC patients with ICIs. For TP53 co-mutation in NSCLC patients, increased TMB and PD-L1 expression were reported, such as TP53 and ATM co-mutation (Ref. Reference Chen119). Furthermore, TP53 and ATM co-mutation were correlated with high TMB regardless of the presence of other driver mutations, such as KRAS (Ref. Reference Chen119). Similarly, in mechanism, co-mutations in DDR way (HRR-MMR or HRR-BER) also presented with increased TMB and neoantigen load and may elevate the expression of immune checkpoint (PD-L1 and LAG3) and T-effector and IFN-γ-associated signatures and so on (Ref. Reference Wang13). Beyond above reports, some co-mutations may exert predictive value independent to TMB. Although emerging studies suggested that high TMB positively correlated with the efficacy of ICIs, Marinelli and coworkers demonstrated that co-mutation of KEAP1-STK11 had a shorter survival in LUAD patients receiving ICIs, even with high TMB, indicating that the level of TMB may not absolutely associated with the efficacy of ICIs (Ref. Reference Marinelli14). Concomitant STK11-KEAP1 loss has been reported to enhance the activation of NRF2 (Ref. Reference Wohlhieter155), which modulates antioxidant response proteins, upregulates ferroptosis-protective mechanisms, and decreases inflammatory factors. Therefore, whether the patients with co-mutation benefit from ICIs may be influenced by various factors, including the interaction between co-mutation genes.

Other mutations were also reported associating with TMB, TIL, and PD-L1 expression. KMT2C was associated with a higher level of TMB and higher PD-L1 expression compared with the wild type due to the genetic instability (Ref. Reference Shi118). MET exon 14 skipping has been reported to have a median TMB burden and demonstrate a poor response to ICIs albeit the higher of PD-L1 expression (Ref. Reference Sabari88) and CD8 + T cell infiltration level (Ref. Reference Li, Wang and Wang87). The co-mutation of MET exon 14 skipping with TP53 will not change the phenomenon. We analysed that the prevalence of co-mutation types of NSCLC patients in TCGA, and summarised the relationship between co-mutation types and their relationship with ICIs therapy (Table 1).

Table 1. The incidence of co-mutations in TCGA and their relationship with ICIs therapy

LUAD, Lung adenocarcinoma; LUSC, Lung squamous cell carcinoma; TCGA, The Cancer Genome Atlas.

Given the relationship between gene mutations and tumour immunity as mentioned, the genetic alterations may influence the TIL in the TME so as to account for tumour immune heterogeneity and immunotherapy sensitivity. The immune-inflamed tumours are recognised as ‘hot tumour’ with high infiltration of CD8 + T lymphocytes, expression of PD-L1, and level of TMB and are more responsive to ICIs (Refs Reference Chen and Mellman9, Reference Chen and Mellman10, Reference Liu and Sun156). While immune-excluded tumours and immune-desert tumours are characterised as ‘cold tumour’ with poor T-cell infiltration or high immunosuppressive cell populations infiltration such as T-regulatory cells (Tregs), myeloid-derived suppressor cells (MDSCs), and tumour-associated macrophages (TAMs) (Ref. Reference Liu and Sun156). For example, EGFR mutation contributes to ‘cold tumour’, characterised as lack of tumour infiltrating lymphocytes (Ref. Reference Dong78) while TP53 and KRAS co-mutation may generate ‘hot tumour’ by increasing T cell infiltration, PD-L1 expression and TMB level (Ref. Reference Dong154). Thus, targeting specific gene mutations or signal pathways may provide us a more insightful perspective to alter tumour immune microenvironment and improve the efficacy of ICIs (Ref. Reference Galon and Bruni157). Meanwhile, gene mutations may also predict the efficacy of ICIs by reconstructing tumour immune microenvironment.

Prospective

In The past, studies demonstrated that NSCLC patients with driver mutations may be less responsive to ICIs than wild type, irrespective of PD-L1 expression. Nonetheless, cancer therapy has a tremendous shift with the advances in sequencing technologies (Ref. Reference Hyman, Taylor and Baselga158). Tumour-specific genetic mutation profile unveils that each patient exists more than one mutation with synergistical or complementary biological changes, indicating that personalised/precision genotype-targeted cancer treatment is imperative (Ref. Reference Martinez-Jimenez2). In addition, immunotherapy intrigues the predominant attention in cancer therapy with the gorgeous advent of several ICIs. The current predictive biomarkers of ICIs have some limitations of PD-L1 expression and TMB level in NSCLC. However, some single-gene mutations have been indicated to correlate with the efficacy of ICIs (such as TP53, KRAS, KEAP1, STK11, BRAF, MET and ALK) or correlated with hyperprogression with anti–PD-1 therapy (such as MDM2/MDM4 amplifications) in NSCLC patients but their predictive ability is influenced by co-occurring mutations and mutation subtype. At the same time, a genomic mutation signature risk model has successfully predicted the efficacy of ICIs in non-squamous NSCLC (Ref. Reference Bai16). Herein, we laid special stress on analysing the recent studies about co-mutations in NSCLC patients, which may be more capable of predicting the effect of ICIs than monogenetic mutations, coping with the dilemma of small groups of immunotherapy response. Given the emergence of co-mutation subtypes, we hope that these specific tumour subtypes will provide better evidence and reference for future clinical decision-making and improve the response rate of ICIs.

We further explored several potential mechanisms among them. Most of the tumour-specific genetic mutations are related to immune-related pathways and T lymphocytes infiltration signalling pathways, such as the secretion of cytokines and the expression of an immune checkpoint. Current researches support that these cellular and molecular properties of lung cancer may construct a vital framework for a patient with immunotherapy. However, more clinical trials with genetical and histological testing are still needed to validate the prognostic effect of co-mutation in immunotherapy by assessing tumour subtype-specific efficacy. With the further study of tumour-specific genetic mutations and its relevant signalling, it is not long before they can be applied to predicting immunotherapy response in patients with NSCLC.

Acknowledgements

We thank Dr Huibo Zhang, Dr Lan Li, Dr Haohan Zhang and Dr Yaqi Zhao of Cancer Center, Renmin Hospital of Wuhan University for their helps in figures preparation.

Financial support

Wuhan University Medical Faculty Innovation Seed Fund Cultivation Project (TFZZ2018025), Chen xiao-ping foundation for the development of science and technology of Hubei province (CXPJJH12000001-2020313) and National Natural Science Foundation of China (grant no. 81670123 and no.81670144).

Conflict of interest

The authors declare no conflict of interest.

Ethical standards

Not applicable.

Footnotes

*

These authors contributed equally.

References

Samadder, NJ et al. (2021) Comparison of universal genetic testing vs guideline-directed targeted testing for patients with hereditary cancer syndrome. JAMA Oncology 7, 230237.CrossRefGoogle ScholarPubMed
Martinez-Jimenez, F et al. (2020) A compendium of mutational cancer driver genes. Nature Reviews Cancer 20, 555572.CrossRefGoogle ScholarPubMed
Thomas, RK et al. (2007) High-throughput oncogene mutation profiling in human cancer. Nature Genetics 39, 347351.CrossRefGoogle ScholarPubMed
Boussiotis, VA (2016) Molecular and biochemical aspects of the PD-1 checkpoint pathway. New England Journal of Medicine 375, 17671778.CrossRefGoogle ScholarPubMed
Lastwika, KJ et al. (2016) Control of PD-L1 expression by oncogenic activation of the AKT-mTOR pathway in non-small cell lung cancer. Cancer Research 76, 227238.CrossRefGoogle ScholarPubMed
Zhang, J et al. (2018) Biochemical aspects of PD-L1 regulation in cancer immunotherapy. Trends in Biochemical Sciences 43, 10141032.CrossRefGoogle ScholarPubMed
Roelands, J et al. (2020) Oncogenic states dictate the prognostic and predictive connotations of intratumoral immune response. Journal for Immunotherapy of Cancer 8, e000617.CrossRefGoogle Scholar
Wu, J et al. (2020) CMTM Family proteins 1–8: roles in cancer biological processes and potential clinical value. Cancer Biology & Medicine 17, 528542.CrossRefGoogle ScholarPubMed
Chen, DS and Mellman, I (2013) Oncology meets immunology: the cancer-immunity cycle. Immunity 39, 110.CrossRefGoogle ScholarPubMed
Chen, DS and Mellman, I (2017) Elements of cancer immunity and the cancer-immune set point. Nature 541, 321330.CrossRefGoogle ScholarPubMed
Zhao, Y et al. (2021) Oncolytic adenovirus: prospects for cancer immunotherapy. Frontiers in Microbiology 12, 707290.CrossRefGoogle ScholarPubMed
Kelly, PN (2018) The cancer immunotherapy revolution. Science 359, 13441345.CrossRefGoogle ScholarPubMed
Wang, Z et al. (2018) Comutations in DNA damage response pathways serve as potential biomarkers for immune checkpoint blockade. Cancer Research 78, 64866496.CrossRefGoogle ScholarPubMed
Marinelli, D et al. (2020) KEAP1-driven co-mutations in lung adenocarcinoma unresponsive to immunotherapy despite high tumor mutational burden. Annals of Oncology: Official Journal of the European Society for Medical Oncology 31, 17461754.CrossRefGoogle ScholarPubMed
Pan, D et al. (2021) A gene mutation signature predicting immunotherapy benefits in patients with NSCLC. Journal of Thoracic Oncology: Official Publication of the International Association for the Study of Lung Cancer 16, 419427.CrossRefGoogle ScholarPubMed
Bai, X et al. (2020) Development and validation of a genomic mutation signature to predict response to PD-1 inhibitors in non-squamous NSCLC: a multicohort study. Journal for Immunotherapy of Cancer 8, e000381.CrossRefGoogle ScholarPubMed
Sun, L et al. (2021) MGA Mutation as a novel biomarker for immune checkpoint therapies in non-squamous non-small cell lung cancer. Frontiers in Pharmacology 12, 625593.CrossRefGoogle ScholarPubMed
Wu, J et al. (2021) A risk model developed based on tumor microenvironment predicts overall survival and associates with tumor immunity of patients with lung adenocarcinoma. Oncogene 40, 44134424.CrossRefGoogle ScholarPubMed
Kumagai, S, Koyama, S and Nishikawa, H (2021) Antitumour immunity regulated by aberrant ERBB family signalling. Nature Reviews Cancer 21, 181197.CrossRefGoogle ScholarPubMed
Siegel, RL, Miller, KD and Jemal, A (2020) Cancer statistics, 2020. CA: a Cancer Journal for Clinicians 70, 730.Google ScholarPubMed
Skoulidis, F and Heymach, JV (2019) Co-occurring genomic alterations in non-small-cell lung cancer biology and therapy. Nature Reviews Cancer 19, 495509.CrossRefGoogle ScholarPubMed
Facchinetti, F et al. (2016) Moving immune checkpoint blockade in thoracic tumors beyond NSCLC. Journal of Thoracic Oncology: Official Publication of the International Association for the Study of Lung Cancer 11, 18191836.CrossRefGoogle ScholarPubMed
Rudd, CE, Taylor, A and Schneider, H (2009) CD28 And CTLA-4 coreceptor expression and signal transduction. Immunological Reviews 229, 1226.CrossRefGoogle ScholarPubMed
Barber, DL et al. (2006) Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 439, 682687.CrossRefGoogle ScholarPubMed
Xia, L, Liu, Y and Wang, Y (2019) PD-1/PD-L1 blockade therapy in advanced non-small-cell lung cancer: current status and future directions. The Oncologist 24, S31S41.CrossRefGoogle ScholarPubMed
Brahmer, J et al. (2015) Nivolumab versus docetaxel in advanced squamous-cell Non-small-cell lung cancer. New England Journal of Medicine 373, 123135.CrossRefGoogle ScholarPubMed
Fehrenbacher, L et al. (2016) Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial. Lancet 387, 18371846.CrossRefGoogle ScholarPubMed
Le, DT et al. (2017) Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 357, 409413.CrossRefGoogle ScholarPubMed
Le, DT et al. (2015) PD-1 Blockade in tumors with mismatch-repair deficiency. New England Journal of Medicine 372, 25092520.CrossRefGoogle ScholarPubMed
Moreira, L et al. (2012) Identification of lynch syndrome among patients with colorectal cancer. JAMA 308, 15551565.CrossRefGoogle ScholarPubMed
Sargent, DJ et al. (2010) Defective mismatch repair as a predictive marker for lack of efficacy of fluorouracil-based adjuvant therapy in colon cancer. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology 28, 32193226.CrossRefGoogle Scholar
Champiat, S et al. (2017) Hyperprogressive disease is a new pattern of progression in cancer patients treated by anti-PD-1/PD-L1. Clinical Cancer Research 23, 19201928.CrossRefGoogle ScholarPubMed
Reck, M et al. (2016) Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. New England Journal of Medicine 375, 18231833.CrossRefGoogle ScholarPubMed
Rittmeyer, A et al. (2017) Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet 389, 255265.CrossRefGoogle ScholarPubMed
Osmani, L et al. (2018) 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. Seminars in Cancer Biology 52, 103109.CrossRefGoogle ScholarPubMed
Bodor, JN, Boumber, Y and Borghaei, H (2020) Biomarkers for immune checkpoint inhibition in non-small cell lung cancer (NSCLC). Cancer 126, 260270.CrossRefGoogle Scholar
Zhou, J et al. (2018) Programmed death ligand 1 expression and CD8(+) tumor-infiltrating lymphocyte density differences between paired primary and brain metastatic lesions in non-small cell lung cancer. Biochemical and Biophysical Research Communications 498, 751757.CrossRefGoogle ScholarPubMed
Yarchoan, M, Hopkins, A and Jaffee, EM (2017) Tumor mutational burden and response rate to PD-1 inhibition. New England Journal of Medicine 377, 25002501.CrossRefGoogle ScholarPubMed
Rizvi, H et al. (2018) Molecular determinants of response to anti-programmed cell death (PD)-1 and anti-programmed death-ligand 1 (PD-L1) blockade in patients with non-small-cell lung cancer profiled with targeted next-generation sequencing. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology 36, 633641.CrossRefGoogle ScholarPubMed
Hellmann, MD et al. (2018) Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden. New England Journal of Medicine 378, 20932104.CrossRefGoogle ScholarPubMed
Hellmann, MD et al. (2018) Genomic features of response to combination immunotherapy in patients with advanced non-small-cell lung cancer. Cancer Cell 33, 843852. e4.CrossRefGoogle ScholarPubMed
Hellmann, MD et al. (2018) Tumor mutational burden and efficacy of Nivolumab monotherapy and in combination with ipilimumab in small-cell lung cancer. Cancer Cell 33, 853861. e4.CrossRefGoogle ScholarPubMed
Assoun, S et al. (2019) Association of TP53 mutations with response and longer survival under immune checkpoint inhibitors in advanced non-small-cell lung cancer. Lung Cancer 132, 6571.CrossRefGoogle ScholarPubMed
Gandara, DR et al. (2017) Blood-based biomarkers for cancer immunotherapy: tumor mutational burden in blood (bTMB) is associated with improved atezolizumab (atezo) efficacy in 2L + NSCLC (POPLAR and OAK). Annals of Oncology 28, v460.CrossRefGoogle Scholar
Cabel, L et al. (2018) Clinical potential of circulating tumour DNA in patients receiving anticancer immunotherapy. Nature Reviews. Clinical Oncology 15, 639650.CrossRefGoogle ScholarPubMed
Guibert, N et al. (2019) Targeted sequencing of plasma cell-free DNA to predict response to PD1 inhibitors in advanced non-small cell lung cancer. Lung Cancer 137, 16.CrossRefGoogle ScholarPubMed
Camidge, DR, Doebele, RC and Kerr, KM (2019) Comparing and contrasting predictive biomarkers for immunotherapy and targeted therapy of NSCLC. Nature Reviews, Clinical Oncology 16, 341355.CrossRefGoogle ScholarPubMed
Rotow, J and Bivona, TG (2017) Understanding and targeting resistance mechanisms in NSCLC. Nature Reviews Cancer 17, 637658.CrossRefGoogle ScholarPubMed
Baluapuri, A, Wolf, E and Eilers, M (2020) Target gene-independent functions of MYC oncoproteins. Nature Reviews Molecular Cell Biology 21, 255267.CrossRefGoogle ScholarPubMed
Cuadrado, A et al. (2019) Therapeutic targeting of the NRF2 and KEAP1 partnership in chronic diseases. Nature Reviews, Drug Discovery 18, 295317.CrossRefGoogle ScholarPubMed
Kotler, E et al. (2018) A systematic p53 mutation library links differential functional impact to cancer mutation pattern and evolutionary conservation. Molecular Cell 71, 178190. e8.CrossRefGoogle ScholarPubMed
Shaw, AT and Engelman, JA (2013) ALK In lung cancer: past, present, and future. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology 31, 11051111.CrossRefGoogle ScholarPubMed
da Cunha Santos, G, Shepherd, FA and Tsao, MS (2011) EGFR Mutations and lung cancer. Annual Review of Pathology 6, 4969.CrossRefGoogle ScholarPubMed
Guertin, DA and Sabatini, DM (2007) Defining the role of mTOR in cancer. Cancer Cell 12, 922.CrossRefGoogle ScholarPubMed
Kastenhuber, ER and Lowe, SW (2017) Putting p53 in context. Cell 170, 10621078.CrossRefGoogle ScholarPubMed
Takaoka, A et al. (2003) Integration of interferon-alpha/beta signalling to p53 responses in tumour suppression and antiviral defence. Nature 424, 516523.CrossRefGoogle ScholarPubMed
Shatz, M, Menendez, D and Resnick, MA (2012) The human TLR innate immune gene family is differentially influenced by DNA stress and p53 status in cancer cells. Cancer Research 72, 39483957.CrossRefGoogle ScholarPubMed
Sun, H et al. (2020) Specific TP53 subtype as biomarker for immune checkpoint inhibitors in lung adenocarcinoma. EBioMedicine 60, 102990.CrossRefGoogle ScholarPubMed
Kato, S et al. (2017) Hyperprogressors after immunotherapy: analysis of genomic alterations associated with accelerated growth rate. Clinical Cancer Research 23, 42424250.CrossRefGoogle ScholarPubMed
Cancer Genome Atlas Research, N (2014) Comprehensive molecular profiling of lung adenocarcinoma. Nature 511, 543550.CrossRefGoogle Scholar
Martin Martorell, P et al. (2017) Coexistence of EGFR, KRAS, BRAF, and PIK3CA mutations and ALK rearrangement in a comprehensive cohort of 326 consecutive Spanish Nonsquamous NSCLC patients. Clinical Lung Cancer 18, e395e402.CrossRefGoogle Scholar
Roman, M et al. (2018) KRAS Oncogene in non-small cell lung cancer: clinical perspectives on the treatment of an old target. Molecular Cancer 17, 33.CrossRefGoogle ScholarPubMed
Scheffler, M et al. (2019) K-ras mutation subtypes in NSCLC and associated co-occuring mutations in other oncogenic pathways. Journal of Thoracic Oncology: Official Publication of the International Association for the Study of Lung Cancer 14, 606616.CrossRefGoogle ScholarPubMed
Hong, DS et al. (2020) KRAS(G12C) inhibition with Sotorasib in advanced solid tumors. New England Journal of Medicine 383, 12071217.CrossRefGoogle ScholarPubMed
Landre, T et al. (2021) Anti-PD-(L)1 for KRAS-mutant advanced non-small-cell lung cancers: a meta-analysis of randomized-controlled trials. Cancer Immunology Immunotherapy 71, 719726.CrossRefGoogle ScholarPubMed
Scheel, AH et al. (2016) PD-L1 expression in non-small cell lung cancer: correlations with genetic alterations. Oncoimmunology 5, e1131379.CrossRefGoogle ScholarPubMed
Yang, Q, Jiang, W and Hou, P (2019) Emerging role of PI3 K/AKT in tumor-related epigenetic regulation. Seminars in Cancer Biology 59, 112124.CrossRefGoogle Scholar
Fruman, DA et al. (2017) The PI3 K pathway in human disease. Cell 170, 605635.CrossRefGoogle Scholar
Alzahrani, AS (2019) PI3 K/Akt/mTOR Inhibitors in cancer: at the bench and bedside. Seminars in Cancer Biology 59, 125132.CrossRefGoogle Scholar
Vazquez, F and Devreotes, P (2006) Regulation of PTEN function as a PIP3 gatekeeper through membrane interaction. Cell Cycle 5, 15231527.CrossRefGoogle ScholarPubMed
Ahmed, SM et al. (2017) Nrf2 signaling pathway: pivotal roles in inflammation. Biochimica et Biophysica Acta, Molecular Basis of Disease 1863, 585597.CrossRefGoogle ScholarPubMed
Hammad, A et al. (2019) “NRF2 addiction” in lung cancer cells and its impact on cancer therapy. Cancer Letters 467, 4049.CrossRefGoogle ScholarPubMed
Singh, A et al. (2021) NRF2 Activation promotes aggressive lung cancer and associates with poor clinical outcomes. Clinical Cancer Research 27, 877888.CrossRefGoogle ScholarPubMed
Best, SA and Sutherland, KD (2018) “Keaping” a lid on lung cancer: the Keap1-Nrf2 pathway. Cell Cycle 17, 16961707.CrossRefGoogle ScholarPubMed
Matsuoka, Y et al. (2016) IL-6 controls resistance to radiation by suppressing oxidative stress via the Nrf2-antioxidant pathway in oral squamous cell carcinoma. British Journal of Cancer 115, 12341244.CrossRefGoogle ScholarPubMed
Shackelford, DB and Shaw, RJ (2009) The LKB1-AMPK pathway: metabolism and growth control in tumour suppression. Nature Reviews Cancer 9, 563575.CrossRefGoogle ScholarPubMed
Koyama, S et al. (2016) STK11/LKB1 Deficiency promotes neutrophil recruitment and proinflammatory cytokine production to suppress T-cell activity in the lung tumor microenvironment. Cancer Research 76, 9991008.CrossRefGoogle ScholarPubMed
Dong, ZY et al. (2017) EGFR Mutation correlates with uninflamed phenotype and weak immunogenicity, causing impaired response to PD-1 blockade in non-small cell lung cancer. Oncoimmunology 6, e1356145.CrossRefGoogle ScholarPubMed
Gainor, JF et al. (2016) EGFR Mutations and ALK rearrangements are associated with low response rates to PD-1 pathway blockade in non-small cell lung cancer: a retrospective analysis. Clinical Cancer Research 22, 45854593.CrossRefGoogle ScholarPubMed
Dankner, M et al. (2018) Classifying BRAF alterations in cancer: new rational therapeutic strategies for actionable mutations. Oncogene 37, 31833199.CrossRefGoogle ScholarPubMed
Negrao, MV et al. (2020) Molecular landscape of BRAF-mutant NSCLC reveals an association between clonality and driver mutations and identifies targetable non-V600 driver mutations. Journal of Thoracic Oncology: Official Publication of the International Association for the Study of Lung Cancer 15, 16111623.CrossRefGoogle ScholarPubMed
Planchard, D et al. (2017) Dabrafenib plus trametinib in patients with previously untreated BRAF(V600E)-mutant metastatic non-small-cell lung cancer: an open-label, phase 2 trial. The Lancet. Oncology 18, 13071316.CrossRefGoogle Scholar
Dudnik, E et al. (2018) BRAF Mutant lung cancer: programmed death ligand 1 expression, tumor mutational burden, microsatellite instability status, and response to immune check-point inhibitors. Journal of Thoracic Oncology: Official Publication of the International Association for the Study of Lung Cancer 13, 11281137.CrossRefGoogle ScholarPubMed
Murciano-Goroff, YR et al. (2021) Immune biomarkers and response to checkpoint inhibition of BRAF(V600) and BRAF non-V600 altered lung cancers. British Journal of Cancer 126, 889898.CrossRefGoogle ScholarPubMed
Trusolino, L, Bertotti, A and Comoglio, PM (2010) MET Signalling: principles and functions in development, organ regeneration and cancer. Nature Reviews Molecular Cell Biology 11, 834848.CrossRefGoogle ScholarPubMed
Awad, MM et al. (2016) 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. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology 34, 721730.CrossRefGoogle ScholarPubMed
Li, X, Wang, R and Wang, L (2020) MET-mutant cancer and immune checkpoint inhibitors: a large database analysis. Lung Cancer 150, 256258.CrossRefGoogle ScholarPubMed
Sabari, JK et al. (2018) PD-L1 expression, tumor mutational burden, and response to immunotherapy in patients with MET exon 14 altered lung cancers. Annals of Oncology: Official Journal of the European Society for Medical Oncology 29, 20852091.CrossRefGoogle ScholarPubMed
Papaccio, F et al. (2018) HGF/MET and the immune system: relevance for cancer immunotherapy. International Journal of Molecular Sciences 19, 3595.CrossRefGoogle ScholarPubMed
Golding, B et al. (2018) The function and therapeutic targeting of anaplastic lymphoma kinase (ALK) in non-small cell lung cancer (NSCLC). Molecular Cancer 17, 52.CrossRefGoogle Scholar
Bakkenist, CJ and Kastan, MB (2003) DNA Damage activates ATM through intermolecular autophosphorylation and dimer dissociation. Nature 421, 499506.CrossRefGoogle ScholarPubMed
Mitui, M et al. (2009) Functional and computational assessment of missense variants in the ataxia-telangiectasia mutated (ATM) gene: mutations with increased cancer risk. Human Mutation 30, 1221.CrossRefGoogle ScholarPubMed
Ali, A (2004) Requirement of protein phosphatase 5 in DNA-damage-induced ATM activation. Genes & Development 18, 249254.CrossRefGoogle ScholarPubMed
Kumar, V, Alt, FW and Oksenych, V (2014) Functional overlaps between XLF and the ATM-dependent DNA double strand break response. DNA Repair (Amst) 16, 1122.CrossRefGoogle ScholarPubMed
Dang, CV (2012) MYC On the path to cancer. Cell 149, 2235.CrossRefGoogle ScholarPubMed
Dang, CV et al. (2006) The c-Myc target gene network. Seminars in Cancer Biology 16, 253264.CrossRefGoogle ScholarPubMed
Chen, H, Liu, H and Qing, G (2018) Targeting oncogenic Myc as a strategy for cancer treatment. Signal Transduct Target Ther 3, 5.CrossRefGoogle ScholarPubMed
Kortlever, RM et al. (2017) Myc cooperates with Ras by programming inflammation and immune suppression. Cell 171, 13011315. e14.CrossRefGoogle ScholarPubMed
Sanchez-Vega, F et al. (2018) Oncogenic signaling pathways in the cancer genome atlas. Cell 173, 321337. e10.CrossRefGoogle ScholarPubMed
Galan-Cobo, A et al. (2019) LKB1 And KEAP1/NRF2 pathways cooperatively promote metabolic reprogramming with enhanced glutamine dependence in KRAS-mutant lung adenocarcinoma. Cancer Research 79, 32513267.CrossRefGoogle ScholarPubMed
Nissan, MH et al. (2014) Loss of NF1 in cutaneous melanoma is associated with RAS activation and MEK dependence. Cancer Research 74, 23402350.CrossRefGoogle ScholarPubMed
Yuan, B et al. (2020) Co-Occurring alterations of ERBB2 exon 20 insertion in non-small cell lung cancer (NSCLC) and the potential indicator of response to Afatinib. Frontiers in Oncology 10, 729.CrossRefGoogle ScholarPubMed
Fang, W et al. (2019) Mutation variants and co-mutations as genomic modifiers of response to Afatinib inHER2-mutant lung adenocarcinoma. The Oncologist 25, e545e554.CrossRefGoogle ScholarPubMed
Mina, M et al. (2017) Conditional selection of genomic alterations dictates cancer evolution and oncogenic dependencies. Cancer Cell 32, 155168. e6.CrossRefGoogle ScholarPubMed
van de Haar, J et al. (2019) Identifying epistasis in cancer genomes: a delicate affair. Cell 177, 13751383.CrossRefGoogle ScholarPubMed
Etemadmoghadam, D et al. (2013) Synthetic lethality between CCNE1 amplification and loss of BRCA1. Proceedings of the National Academy of Sciences of the USA 110, 1948919494.CrossRefGoogle ScholarPubMed
Brown, JS et al. (2017) Targeting DNA repair in cancer: beyond PARP inhibitors. Cancer Discovery 7, 2037.CrossRefGoogle ScholarPubMed
George, A, Kaye, S and Banerjee, S (2017) Delivering widespread BRCA testing and PARP inhibition to patients with ovarian cancer. Nature Reviews, Clinical Oncology 14, 284296.CrossRefGoogle ScholarPubMed
Carey, JPW et al. (2018) Synthetic lethality of PARP inhibitors in combination with MYC blockade is independent of BRCA Status in triple-negative breast cancer. Cancer Research 78, 742757.CrossRefGoogle ScholarPubMed
Yang, W et al. (2020) Favorable immune microenvironment in patients with EGFR and MAPK Co-mutations. Lung Cancer: Targets and Therapy 11, 5971.Google ScholarPubMed
Skoulidis, F et al. (2018) STK11/LKB1 Mutations and PD-1 inhibitor resistance in KRAS-mutant lung adenocarcinoma. Cancer Discovery 8, 822835.CrossRefGoogle ScholarPubMed
Peng, DH et al. (2021) Th17 cells contribute to combination MEK inhibitor and anti-PD-L1 therapy resistance in KRAS/p53 mutant lung cancers. Nature Communications 12, 2606.CrossRefGoogle ScholarPubMed
Arbour, KC et al. (2018) Effects of co-occurring genomic alterations on outcomes in patients with KRAS-mutant non-small cell lung cancer. Clinical Cancer Research 24, 334340.CrossRefGoogle ScholarPubMed
Frost, N et al. (2021) KRAS(G12C)/TP53 co-mutations identify long-term responders to first line palliative treatment with pembrolizumab monotherapy in PD-L1 high (≥50%) lung adenocarcinoma. Translational Lung Cancer Research 10, 737752.CrossRefGoogle ScholarPubMed
Gao, G et al. (2020) KRAS G12D mutation predicts lower TMB and drives immune suppression in lung adenocarcinoma. Lung Cancer 149, 4145.CrossRefGoogle ScholarPubMed
Best, SA et al. (2018) Synergy between the KEAP1/NRF2 and PI3 K pathways drives non-small-cell lung cancer with an altered immune microenvironment. Cell Metabolism 27, 935943. e4.CrossRefGoogle Scholar
Jiang, T et al. (2021) Toripalimab plus chemotherapy as second-line treatment in previously EGFR-TKI treated patients with EGFR-mutant-advanced NSCLC: a multicenter phase-II trial. Signal Transduct Target Ther 6, 355.CrossRefGoogle ScholarPubMed
Shi, Y et al. (2021) Integration of comprehensive genomic profiling, tumor mutational burden, and PD-L1 expression to identify novel biomarkers of immunotherapy in non-small cell lung cancer. Cancer Medicine 10, 22162231.CrossRefGoogle ScholarPubMed
Chen, Y et al. (2019) Association of tumor protein p53 and ataxia-telangiectasia mutated comutation with response to immune checkpoint inhibitors and mortality in patients with non-small cell lung cancer. JAMA Netw Open 2, e1911895.CrossRefGoogle ScholarPubMed
Mazzotta, M et al. (2020) Efficacy of immunotherapy in lung cancer with co-occurring mutations in NOTCH and homologous repair genes. Journal for Immunotherapy of Cancer 8, e000946.CrossRefGoogle ScholarPubMed
Scarbrough, PM et al. (2016) A cross-cancer genetic association analysis of the DNA repair and DNA damage signaling pathways for lung, ovary, prostate, breast, and colorectal cancer. Cancer Epidemiology, Biomarkers & Prevention: A Publication of the American Association for Cancer Research, Cosponsored by the American Society of Preventive Oncology 25, 193200.CrossRefGoogle ScholarPubMed
Chang, A et al. (2021) Recruitment of KMT2C/MLL3 to DNA damage sites mediates DNA damage responses and regulates PARP inhibitor sensitivity in cancer. Cancer Research 81, 33583373.CrossRefGoogle ScholarPubMed
Zhang, P and Huang, Y (2021) Genomic alterations in KMT2 family predict outcome of immune checkpoint therapy in multiple cancers. Journal of Hematology & Oncology 14, 39.CrossRefGoogle ScholarPubMed
Wong, SK et al. (2021) MET Exon 14 skipping mutation positive non-small cell lung cancer: response to systemic therapy. Lung Cancer 154, 142145.CrossRefGoogle ScholarPubMed
Albitar, M et al. (2018) Correlation of MET gene amplification and TP53 mutation with PD-L1 expression in non-small cell lung cancer. Oncotarget 9, 1368213693.CrossRefGoogle ScholarPubMed
Labbe, C et al. (2017) Prognostic and predictive effects of TP53 co-mutation in patients with EGFR-mutated non-small cell lung cancer (NSCLC). Lung Cancer 111, 2329.CrossRefGoogle Scholar
Zheng, C et al. (2020) Coexisting EGFR and TP53 mutations in lung adenocarcinoma patients are associated with COMP and ITGB8 upregulation and poor prognosis. Frontiers in Molecular Biosciences 7, 30.CrossRefGoogle ScholarPubMed
Zhao, Y et al. (2020) EGFR-mutant lung adenocarcinoma harboring co-mutational tumor suppressor genes predicts poor prognosis. Journal of Cancer Research and Clinical Oncology 146, 17811789.CrossRefGoogle ScholarPubMed
Gu, M, Xu, T and Chang, P (2021) KRAS/LKB1 and KRAS/TP53 co-mutations create divergent immune signatures in lung adenocarcinomas. Therapeutic Advances in Medical Oncology 13, 17588359211006950.CrossRefGoogle ScholarPubMed
Wang, L et al. (2014) PIK3CA Mutations frequently coexist with EGFR/KRAS mutations in non-small cell lung cancer and suggest poor prognosis in EGFR/KRAS wildtype subgroup. PLoS ONE 9, e88291.CrossRefGoogle ScholarPubMed
Lee, CK et al. (2017) Checkpoint inhibitors in metastatic EGFR-mutated non-small cell lung cancer-A meta-analysis. Journal of Thoracic Oncology: Official Publication of the International Association for the Study of Lung Cancer 12, 403407.CrossRefGoogle ScholarPubMed
Cho, JW et al. (2021) Dysregulation of TFH-B-TRM lymphocyte cooperation is associated with unfavorable anti-PD-1 responses in EGFR-mutant lung cancer. Nature Communications 12, 6068.CrossRefGoogle ScholarPubMed
Chen, N et al. (2017) KRAS mutation-induced upregulation of PD-L1 mediates immune escape in human lung adenocarcinoma. Cancer Immunology Immunotherapy 66, 11751187.CrossRefGoogle ScholarPubMed
Shamalov, K et al. (2017) The mutational status of p53 can influence its recognition by human T-cells. Oncoimmunology 6, e1285990.CrossRefGoogle ScholarPubMed
Topper, MJ et al. (2017) Epigenetic therapy ties MYC depletion to reversing immune evasion and treating lung cancer. Cell 171, 12841300. e21.CrossRefGoogle ScholarPubMed
Parkes, EE et al. (2017) Activation of STING-dependent innate immune signaling by S-phase-specific DNA damage in breast cancer. Journal of the National Cancer Institute 109, djw199.CrossRefGoogle ScholarPubMed
Thiem, A et al. (2019) IFN-gamma-induced PD-L1 expression in melanoma depends on p53 expression. Journal of Experimental & Clinical Cancer Research: CR 38, 397.CrossRefGoogle ScholarPubMed
Lei, TY et al. (2021) The immune response of T cells and therapeutic targets related to regulating the levels of T helper cells after ischaemic stroke. Journal of Neuroinflammation 18, 25.CrossRefGoogle ScholarPubMed
Nogami, N et al. (2021) IMPower150 final exploratory analyses for Atezolizumab Plus bevacizumab and chemotherapy in key NSCLC patient subgroups with EGFR mutations or metastases in the liver or brain. Journal of Thoracic Oncology 17, 309323.CrossRefGoogle ScholarPubMed
Peng, S et al. (2019) EGFR-TKI resistance promotes immune escape in lung cancer via increased PD-L1 expression. Molecular Cancer 18, 165.CrossRefGoogle ScholarPubMed
Matsuzaki, S et al. (2010) Lysophosphatidic acid inhibits CC chemokine ligand 5/RANTES production by blocking IRF-1-mediated gene transcription in human bronchial epithelial cells. Journal of Immunology 185, 48634872.CrossRefGoogle ScholarPubMed
Coelho, MA et al. (2017) Oncogenic RAS signaling promotes tumor immunoresistance by stabilizing PD-L1 mRNA. Immunity 47, 10831099. e6.CrossRefGoogle ScholarPubMed
Barber, GN (2015) STING: infection, inflammation and cancer. Nature Reviews Immunology 15, 760770.CrossRefGoogle Scholar
Hartlova, A et al. (2015) DNA Damage primes the type I interferon system via the cytosolic DNA sensor STING to promote anti-microbial innate immunity. Immunity 42, 332343.CrossRefGoogle ScholarPubMed
Zhang, Y et al. (2021) MET Amplification attenuates LungTumor response to immunotherapy by inhibiting STING. Cancer Discovery 11, 27262737.CrossRefGoogle Scholar
Schabath, MB et al. (2016) Differential association of STK11 and TP53 with KRAS mutation-associated gene expression, proliferation and immune surveillance in lung adenocarcinoma. Oncogene 35, 32093216.CrossRefGoogle ScholarPubMed
Kitajima, S et al. (2019) Suppression of STING associated with LKB1 loss in KRAS-driven lung cancer. Cancer Discovery 9, 3445.CrossRefGoogle ScholarPubMed
Schumacher, TN and Schreiber, RD (2015) Neoantigens in cancer immunotherapy. Science 348, 6974.CrossRefGoogle ScholarPubMed
Zhao, L et al. (2020) TP53 Somatic mutations are associated with poor survival in non-small cell lung cancer patients who undergo immunotherapy. Aging (Albany NY) 12, 1455614568.CrossRefGoogle ScholarPubMed
Cha, YJ et al. (2016) Clinicopathological and prognostic significance of programmed cell death ligand-1 expression in lung adenocarcinoma and its relationship with p53 status. Lung Cancer 97, 7380.CrossRefGoogle ScholarPubMed
Cortez, MA et al. (2016) PDL1 Regulation by p53 via miR-34. Journal of the National Cancer Institute 108, djv303.CrossRefGoogle ScholarPubMed
Singal, G et al. (2019) Association of patient characteristics and tumor genomics with clinical outcomes among patients with non-small cell lung cancer using a clinicogenomic database. JAMA 321, 13911399.CrossRefGoogle ScholarPubMed
Gettinger, S and Politi, K (2016) PD-1 Axis inhibitors in EGFR- and ALK-driven lung cancer: lost cause? Clinical Cancer Research: an Official Journal of the American Association for Cancer Research 22, 45394541.CrossRefGoogle ScholarPubMed
Dong, ZY et al. (2017) Potential predictive value of TP53 and KRAS mutation status for response to PD-1 blockade immunotherapy in lung adenocarcinoma. Clinical Cancer Research 23, 30123024.CrossRefGoogle ScholarPubMed
Wohlhieter, CA et al. (2020) Concurrent mutations in STK11 and KEAP1 promote ferroptosis protection and SCD1 dependence in lung cancer. Cell Reports 33, 108444.CrossRefGoogle ScholarPubMed
Liu, YT and Sun, ZJ (2021) Turning cold tumors into hot tumors by improving T-cell infiltration. Theranostics 11, 53655386.CrossRefGoogle ScholarPubMed
Galon, J and Bruni, D (2019) Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nature Reviews, Drug Discovery 18, 197218.CrossRefGoogle ScholarPubMed
Hyman, DM, Taylor, BS and Baselga, J (2017) Implementing genome-driven oncology. Cell 168, 584599.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. The downstream signalling pathway of Tumour-related gene mutation. Bold fonts indicated that the protein existed corresponding gene mutation. Green represented suppressor gene while orange represented proto-oncogene.

Figure 1

Fig. 2. The immune microenvironment of tumour. The tumour immune microenvironment changed with the onset and evolution of tumour. Tumour antigen was presented to CD8+ T cells directly or presented to CD4+ T cells indirectly by APC (antigen-presenting cell) to elicit anti-tumour response. M1 macrophage cells also exerted anti-tumour immune response. Additionally, tumour cells also secreted chemokines such as CXCL10 to recruit lymphocytes. Tumour-associated macrophage, Tregs and MDSC play a role in suppressing anti-tumour immune response through secreting TGF-β, IL-10, and expressing IDO-1 and IDO-2. Neutrophils secreted neutrophil elastase (NE) to promote tumour proliferation. Meanwhile, cellular matrix mediated by TGF-β in tumour blocked the infiltration of lymphocytes. Cancer-associated fibroblasts (CAFs) released stromal cell derived factors, angiogenic factors to promote tumour cell growth and tumour blood vessel regeneration.

Figure 2

Fig. 3. The pathway of gene mutation to influence the tumour-associated immune response. The diverse signalling pathways and transcriptional factors promoted the expression of PD-L1 to assist tumour to evade immune response. Tumour cells evade anti-tumour effects of T cells in part by elevating PD-L1 mRNA expression at transcriptional level via activation of different upstream signalling pathways.

Figure 3

Table 1. The incidence of co-mutations in TCGA and their relationship with ICIs therapy