Skip to main content
Erschienen in: Clinical and Experimental Medicine 1/2024

Open Access 01.12.2024 | Review

Multi-omics and artificial intelligence predict clinical outcomes of immunotherapy in non-small cell lung cancer patients

verfasst von: Ting Mei, Ting Wang, Qinghua Zhou

Erschienen in: Clinical and Experimental Medicine | Ausgabe 1/2024

Abstract

In recent years, various types of immunotherapy, particularly the use of immune checkpoint inhibitors targeting programmed cell death 1 or programmed death ligand 1 (PD-L1), have revolutionized the management and prognosis of non-small cell lung cancer. PD-L1 is frequently used as a biomarker for predicting the likely benefit of immunotherapy for patients. However, some patients receiving immunotherapy have high response rates despite having low levels of PD-L1. Therefore, the identification of this group of patients is extremely important to improve prognosis. The tumor microenvironment contains tumor, stromal, and infiltrating immune cells with its composition differing significantly within tumors, between tumors, and between individuals. The omics approach aims to provide a comprehensive assessment of each patient through high-throughput extracted features, promising a more comprehensive characterization of this complex ecosystem. However, features identified by high-throughput methods are complex and present analytical challenges to clinicians and data scientists. It is thus feasible that artificial intelligence could assist in the identification of features that are beyond human discernment as well as in the performance of repetitive tasks. In this paper, we review the prediction of immunotherapy efficacy by different biomarkers (genomic, transcriptomic, proteomic, microbiomic, and radiomic), together with the use of artificial intelligence and the challenges and future directions of these fields.
Hinweise
Ting Mei, Ting Wang these authors have contributed equally to this work.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Lung cancer is responsible for the majority of cancer-related deaths in the world. Non-small cell lung cancer (NSCLC) represents approximately 80–90% of lung cancers [1]. Over half of NSCLC tumors are advanced when diagnosed, and the overall 5-year survival for NSCLC patients is only 18% [2].
In recent years, the use of various types of immunotherapy, particularly immune checkpoint inhibitors (ICIs) targeting programmed cell death 1 (PD-1) or programmed death ligand 1 (PD-L1), has revolutionized the treatment of NSCLC [3, 4]. Both PD-1 and PD-L1 are important immune checkpoints (ICs). PD-1 is expressed on the surfaces of immune cells and binds its ligand, PD-L1, expressed on tumor cells, suppressing the immune response against the tumor [5]. The function of anti-PD-1/PD-L1 immunotherapy is to prevent this interaction and thus enhance the immune response directed against the tumor [6].
Immunohistochemical (IHC) detection of PD-L1 has FDA approval and is the most frequently used biomarker for the prediction of the effectiveness of immunotherapy [7]. However, anomalous results have been observed in clinical practice, with many patients with high PD-L1 levels showing a poor response to immunotherapy while others with low or negative PD-L1 levels do respond. Furthermore, the efficacy of immunotherapy guided by PD-L1 expression levels does not exceed 30% [8]. This suggests that the use of PD-L1 as a sole biomarker is inadequate.
The tumor microenvironment (TME) contains various cell types, including tumor, stromal, and infiltrating immune cells, and its composition differs significantly within tumors [9], between tumor types [10], and among individuals [11]. In such a heterogeneous system, many pro- and anti-tumor cellular components or signals can influence the efficacy of immunotherapy. Thus, the tumor response to immunotherapy is complex and involves a large number of mechanisms and pathways. Omics approaches, aimed at the comprehensive assessment of each patient through the analysis of high-throughput-extracted features, promise to provide a more comprehensive characterization of this complex ecosystem.
Artificial intelligence (AI) is a subdivision of computer science that can be used to predict and classify material according to the available data [12]. Deep learning (DL) is a representative learning method in which complex multi-layer neural network architectures automatically learn from data by converting input information into multi-level abstractions. At the same time, models are constructed by the selection of features and model fitting using a combination of computerized algorithms and high-throughput data. Previous studies have shown that AI-based automatic learning and diagnostic models have shown good performance in cell classification [13], cancer detection [14], pathological diagnosis [15], and the analysis of multiple biomarkers [16].
Here, we review the use of multi-omics (genomics, transcriptomics, proteomics, microbiomics, and radiomics) for the prediction of the outcome of immunotherapy and specifically discuss the applications of AI in these predictions (Fig. 1).

Genomics

Tumor mutation burden

The TMB represents the total number of somatic mutations present in the tumor tissue after the exclusion of germline mutations. Somatic mutations can produce changes in cell surface proteins, leading to the appearance of neoantigens that are recognized as foreign by the immune system, stimulating the immune response and subsequent elimination of the tumor cells [17]. There is, thus, a significant association between high TMB and the patient’s response to immunotherapy.
In the CheckMate 026 study, an exploratory analysis of the influence of tumor mutational burden (TMB) on clinical outcome showed that in patients with high TMB levels (≥ 243 mutations/Mb), those receiving immunotherapy had longer PFS (9.7 months vs. 5.8 months for chemotherapy, HR, 0.62 (95%CI, 0.38–1.00)) and higher objective response rates (ORRs) (46.8% vs. 28.3% for chemotherapy) [18]. The Checkmate 227 study used TMB ≥ 10 mut/Mb as the cutoff value for high TMB and found markedly improved ORR and PFS when nivolumab was combined with Ipilimumab compared with chemotherapy in advanced NSCLC patients with high TMB, regardless of PD-L1 expression level [19]. These two studies suggest that the TMB may be useful as a complementary biomarker for the selection of patients likely to respond to immunotherapy. Next, the bFAST cohort C study and a multi-center retrospective study set the TMB threshold at 16 mutations/Mb and 20 mutations/Mb, respectively, and also found that high TMB was associated with better patient outcomes [20, 21]. However, there is a problem that these studies have inconsistent definitions of high TMB, and the inconsistencies in TMB detection platforms and methods, sample status, and result interpretation further interfere with the prediction of clinical outcomes. Therefore, TMB requires synchronization and standardization for reliable clinical application. In addition, it is worth noting that TMB still has serious limitations as a predictive factor in immunotherapy combined with chemotherapy.

Circulating tumor DNA (ctDNA)

Measurement of ctDNA in the peripheral blood can provide an accurate reflection of the TMB and genomics of the cancer and can thus be used as a biomarker predicting treatment response and prognosis.
Assessment of ctDNA levels before treatment has been demonstrated to be useful for outcome prediction, with high ctDNA scores indicative of poor outcomes in many cancer types [22, 23]. A prospective phase II clinical trial observed an association between lower baseline ctDNA levels and enhanced OS and PFS after treatment of NSCLC with pembrolizumab [24]. Several recent studies have shown an independent association between early reduction in the ctDNA allele frequency and enhanced OS and ORR in patients with advanced NSCLC after immunotherapy [24, 25]. Vokes et al. reported at the 2023 American Society of Clinical Oncology (ASCO) that dynamic changes in ctDNA can predict the therapeutic efficacy of cemiplimab [26]. Pseudoprogression can also be identified by ctDNA kinetics. For instance, a study by Guibert et al. observed rapid and significantly reduced ctDNA levels in two patients with pseudoprogression while patients with true progression showed increased levels [27].
Although ctDNA levels are considered predictors of response to immunotherapy, there is no uniform standard for baseline risk stratification thresholds and time points for testing. Therefore, further cross-study validations and standardization of ctDNA thresholds are needed in the future.

Genetic mutations

EGFR mutations

Together with the total mutational burden, there are specific mutations that may influence the efficacy of immunotherapy. Meta-analyses of three clinical trials (CheckMate 057, POPLAR, and KEYNOTE 010) and meta-analyses of five trials (CheckMate 017, CheckMate 057, KEYNOTE 010, OAK, and POPLAR) confirmed that single-agent ICI does not prolong OS in patients with mutations in EGFR [28, 29]. The CheckMate 012 study found that immunotherapy combined with chemotherapy similarly failed to improve PFS and OS in patients with EGFR mutations [30]. However, the ORIENT-31 study confirmed that ICI combined with anti-angiogenic therapy and chemotherapy significantly improved PFS in EGFR-mutant non-squamous NSCLC patients who progressed after treatment with EGFR-TKIs [31]. Although the IMpower150 [32], ORIENT-31 and ATTLAS/KCSG-LU19-04 [33] studies have unanimously confirmed that patients with EGFR-TKI-resistant NSCLC can benefit from the four-drug combination therapy of immune + platinum-containing double-drug chemotherapy + bevacizumab. However, the four-drug combination regimen has a higher incidence of adverse effects and is limited in clinical use. Checkmate 722 and Keynote 789 studies chose immunotherapy + platinum-based doublet chemotherapy, but the efficacy was not satisfactory. The Checkmate 722 study demonstrated that nivolumab + chemotherapy had no significant clinical benefit compared with chemotherapy in patients with EGFR-TKI resistance [34]. In the Keynote 789 study, pembrolizumab repeated the failed results [35]. Whether other immune drugs combined with chemotherapy are effective, and whether immunotherapy + anti-angiogenic therapy + single-agent chemotherapy is feasible and is worthy of further exploration in the future.

ALK or ROS1 fusion-positive

NSCLC patients with the ALK/ROS1 fusion are classified as having low TMB despite their high levels of PD-L1 expression [36]. The ATLANTIC study has shown that the ORR of patients with ALK rearrangements after immunotherapy was significantly lower than that of ALK-negative patients [37]. The results of the IMMUNOTARGET study found that the ORRs of NSCLC patients with positive ALK and ROS1 driver genes treated with ICIs were 0% and 17%, respectively [38].

RET rearrangement

Patients with NSCLC accompanied by RET rearrangements also typically have lower TMB levels (< 2.5 mut/Mb) and PD-L1 expression [36, 39]. Previous retrospective studies have shown that the curative effect of ICI therapy in patients with RET-rearranged NSCLC is poor, with a median PFS of < 3.5 months and an ORR of < 10% [40, 41].

HER2 mutations

Marcelo et al. analyzed data from more than 4000 driver-positive NSCLC patients, observing that patients with classic EGFR mutation and HER2 mutations who were treated with ICIs had the shortest PFS (1.8 and 1.9 months, respectively) [36]. The POLISH study also found that for advanced NSCLC patients with HER2 mutations, chemotherapy + immunotherapy did not improve median PFS compared with chemotherapy + anti-angiogenic therapy or chemotherapy alone (both p > 0.05) [42].

KRAS mutations

Mutations in KRAS are often associated with both a high TMB and significantly elevated PD-L1 expression [43]. Moreover, previous studies found no significant differences in ORR, PFS, and OS between NSCLC patients with or without KRAS mutations [44, 45].
Mutations in KRAS are highly heterogeneous. Ricciuti et al. found that among patients with KRAS mutations receiving immunotherapy, those with the KRAS G12D mutation had worse ORR, PFS, and OS than patients with other KRAS mutations [46].
KRAS mutations in NSCLC are frequently seen in conjunction with mutations in tumor protein 53 (TP53) and serine-threonine kinase 11/liver kinase B1 (STK11/LKB1). These KRAS-STK11/LKB1 co-mutant tumors tended to show greater resistance to PD-1 inhibitors [47], while the patients with the KRAS-TP53 co-mutation were more sensitive to PD-1 inhibitors [48]. Lung cancer patients with KRAS mutations have a 20% and 10% chance of co-occurring KEAP1 and SMARCA4 mutations, respectively [49, 50], and these co-mutations appear to be linked to poorer outcomes after immunotherapy.

BRAF mutations

BRAF-mutant NSCLC often has a high TMB and high levels of PD-L1 expression [36, 51]. Li et al. found no significant difference in PFS, OS, and ORR between NSCLC patients carrying BRAF-mutant or wild-type genes after single-agent ICI therapy or ICI combination therapy [52]. A study observed shorter PFS and OS in patients receiving ICIs in the patients carrying the BRAF V600E mutation compared with those without the mutation [53].

MET exon 14

Studies by Negrao et al. and Mazieres et al. found that NSCLC patients with MET exon 14 mutations who were treated with ICIs had a median PFS of < 5 months and an ORR of < 20% [36, 38]. Similarly, Sabari et al. found that the overall response to ICIs was reduced in NSCLC patients with MET exon 14 mutations regardless of PD-L1 expression and the TMB level [54].

Transcriptomics

Transcriptomics involves the analysis of all RNA transcripts within a cell type and is usually performed using high-throughput techniques such as RNA sequencing (RNA-seq) and microarrays [55]. A number of recent studies have investigated the prediction of immunotherapy response using transcriptome signatures.
Ayers analyzed gene expression profiling (GEP) using RNA from baseline tumor samples from patients treated with pembrolizumab, identifying the GEPs of T-cell inflammation, specifically, genes involved in antigen presentation after IFN-γ activation, the expression of chemokines, cytotoxicity, and resistance to the adaptive immune response [56]. Another clinical trial involving 20 groups of patients with advanced solid tumors further validated the relationship between T-cell inflammatory GEP and the clinical efficacy of pembrolizumab and found that tumors with higher T-cell inflammatory GEP showed higher response rates and longer PFS [57]. Further analysis by the POPLAR study showed that among patients treated with atezolizumab, patients with high expression of T effector-interferon-γ-related genes had significantly longer OS [58]. In addition, in NSCLC patients receiving durvalumab, those with a four-gene IFNγ-positive (IFNγ +) signature, namely IFNγ, CD274, LAG3, and CXCL9, showed greater ORR and longer median OS and PFS, irrespective of their PD-L1 status [59]. These transcriptomic data can help to further optimize cancer immunotherapy strategies. However, these studies have tended to analyze specific genes in isolation, and the underlying pathways through which the genes influence the efficacy of ICI therapy require further investigation.

Proteomics

Protein is one of the main building blocks of cells. Proteomics is the large-scale study of proteins through analysis of their identities and quantities in biological samples (cells, tissues, or body fluids).
Using spatially resolved proteomic analysis, Myrto et al. identified tumor cell CD44 as a biomarker for the prediction of the efficacy of immunotherapy in NSCLC patients [60]. Shen et al. analyzed SEL1L3 in a large-scale tissue proteomics dataset established by the Taiwan Cancer Moon Shot Program and found that it could serve as a potential emerging adjuvant for immunotherapy of lung adenocarcinoma [61]. To verify the real performance of blood-based proteomic analysis in NSCLC immunotherapy, all patients were tested and designated as HIC-Hot (HIC-H) or HIC-Cold (HIC-C) before initiating treatment. The study found that the survival time of all HIC-H patients was significantly longer than that of HIC-C patients, irrespective of PD-L1 expression. Additionally, the data also suggest that HIC-C patients should not be treated with ICIs alone, regardless of PD-L1 expression [62]. Furthermore, the combination of protein markers (CXCL9, CXCL10, IL-15, CASP8, and ADA) was found to be more accurate in predicting response than tumor PD-L1 expression or the levels of individual proteins [63].
Notably, although proteomic studies are able to predict the immunotherapy response, there are still some limitations. These tests are based on protein signatures, and the quality of the signature is determined by the quality of the data used in the signature generation. In addition, proteomic analyses based on tumor blood typically measure proteins shed from the tumor into the blood. Thus, the effectiveness of these tests is dependent on the amount of shedding, which is likely to vary between patients. Finally, most studies focusing on proteomics-based biomarkers have been retrospective, and as such they lack informed treatment guidelines, such as NCCN guidelines. Thus, more prospective and randomized clinical trials are required in this area.

Microbiomics

The microbiome refers to the colonies of symbiotic microorganisms within the human body. Analysis of the microbiome is also thought to be able to predict the response to ICI treatment.

Gut microbiota

A study published by Routy in 2018 showed that antibiotic treatment reduced the OS of patients with advanced NSCLC by half within 3 months of immunotherapy [64]. Significant differences in fecal microbiomes were observed before the start of ICI treatment between patients who responded to treatment and those who did not, with Akkermansia muciniphila and Enterococcus hirae found to be significantly related to improved clinical outcomes. Routy et al. reported that ICI resistance could be reduced in mice by transplantation of Akkermansia muciniphila and Enterococcus hirae, suggesting that the gut microbiome modulates the effectiveness of immunotherapy and that ICI efficacy is related to gut bacterial type and can improve immune tolerance in patients by supplementing with beneficial bacteria. A multi-center prospective observational study in 2019 further verified that among NSCLC patients treated with ICI, those with higher levels of Akkermansia muciniphila showed better response rates [65].
These studies demonstrated that the microbiome composition differs according to treatment response, and while favorable results were consistently associated with increased microbiome diversity, differences were observed in the individual bacteria between studies. These differences may be due to the type of ICI used, and geographical location may also play a role.

Local tumor microbiome

The gut microbiota has been linked to the therapeutic effectiveness of immunotherapy through a variety of mechanisms. For lung cancer, local tumor-microbiome interactions are also crucial for the efficacy and prognosis of ICI therapy. The results of Brutsche et al. showed that, as with gut microbes, more diverse bacterial metagenomes were linked to improved survival after ICI treatment. The presence of Gammaproteobacteria has been associated with reduced PD-L1 levels and decreased response to immunotherapy [66]. Sang Hoon Lee et al. observed that V. dispar was more abundant in patients that responded to immunotherapy while Haemophilus influenzae and Neisseria perflava were more abundant in patients who did not. Thus, Haemophilus may represent a target for lung cancer treatment [67]. However, mechanistic data on the function of the microbiome in relation to NSCLC immunotherapy are scarce at this stage, and further investigation is required.
Both preclinical and clinical studies have provided insight into the influence of the microbiome on the immunotherapy response. Future studies should address the classification of favorable and unfavorable characteristics of the microbiota and their influence on the cells and pathways of the immune response to provide a full understanding of their roles in TMEs and to enhance the effectiveness of treatment. The gut-pulmonary axis refers to the commonality between the microbiomes of the lung and gut which are not separate entities but rather communicate in a bidirectional manner to modulate each other's immune responses. The gut-pulmonary axis offers the possibility of indirect modulation of the lung microbiota by manipulating the more accessible gut microbiota.

Radiomics

Radiomics converts medical images into quantitative data, dissecting the complexity of tumor biology both macroscopically and microscopically. Radiomics offers a more comprehensive analysis of the tumor than analysis of biopsy specimens as it extracts features from the entire TME. Radiomic models can also be applied in multiple scans, allowing the non-invasive and continuous monitoring of the tumor and its response to treatment.

Prediction of immunotherapy efficacy

Tunali et al. analyzed pre-immunotherapy data from NSCLC patients treated with PD-1/PD-L1 from 13 institutions, finally identifying four radiomic features that were used for the construction of a model for the differentiation of patient response to ICI with an AUC of 0.790 [68]. Lu et al. selected 88 radiomic features from CT images to build a random forest model and combined clinicopathological factors to determine the patients that were most likely to respond to immunotherapy before beginning treatment (AUC: 0.848–0.902) [69]. Using a combination of CT images and genomic data, Sun et al. developed a radiomic signature of CD8 cells that were effective in predicting treatment outcomes with anti-PD-1 or anti-PD-L1 immunotherapy [70].

Identification of hyperprogression and pseudoprogression

Hyperprogression describes the apparent paradox of increased progression occurring after the initiation of immunotherapy. Patients who experience hyperprogression have a poor prognosis, and early identification of hyperprogression may prevent ineffective treatment and reduce the occurrence of toxicity. Pranjal Vaidya et al. extracted a total of 198 radiomic texture patterns as well as nodule-associated vascular features from CT scans for modeling to identify hyperprogression, demonstrating AUCs of 0.850 and 0.960 for the training and validation sets, respectively [71]. In addition, Tunali et al. combined radiomic models with clinical features and found relatively high predictive power for hyperprogression, with AUCs ranging from 0.804 to 0.865 [72]. In addition, distinguishing pseudoprogression from true disease progression is important for the clinical decision-making process. Barabino et al. found that changes in nine radiomic features were significantly associated with pseudoprogression [73].
These studies show that while radiomics can be a better predictor of ICI treatment efficacy, several problems remain, including a lack of high-quality data and image standardization, together with a poorly effective combination of multiple imaging methods, and a lack of prospective validation. In future studies, a variety of modeling approaches should be used for the selection of the best method. Furthermore, multiple cross-validations should be conducted for single-center studies. In conclusion, we consider that the use of radiomics in PD-L1-related imaging of NSCLC patients has potential.

The application of AI in predicting the response to immunotherapy

AI-based genomics

He et al. used 1020 DL features to build a model to distinguish between patients with high and low TMB, finding that the model was effective in dividing patients into high-risk and low-risk groups when predicting the efficacy of immunotherapy [74]. Jain and Massoud combined three DL models to predict TMB status, reporting an AUC of the joint model of 0.920 [75]. These studies demonstrate the value of an AI system to assist healthcare professionals in identifying the expression status of these genes that have the advantages of being easy to use and non-invasive.

AI-based transcriptomics

Intratumoral heterogeneity (ITH) is a key factor influencing the patient response to immunotherapy [76]. To determine ITH levels, Sung et al. used an ML-based approach to predict ITH using transcriptomic data. They demonstrated that their method could distinguish tumor samples with high and low ITH levels and identify transcriptomic markers associated with ITH [77]. To facilitate the investigation of heterogeneity in the ICI response, Zeng et al. developed a non-negative matrix factorization (NMF)-based ML framework to identify factors affecting the immunotherapy response using data from TCGA samples [78]. With the advent of the era of precision medicine, AI-assisted transcriptomic technology will continue to develop and become more common in immunotherapy research, providing valuable information for the formulation of new strategies to improve the efficacy of cancer immunotherapy.

AI-based proteomics

A study reported the construction of an ML-based serum protein classifier that could classify patients into drug-resistant, intermediate, or sensitive groups according to clinical and mass spectrometry (MS) characteristics. This serum protein classifier showed good performance in predicting patient immune responses [79]. The VeriStrat test is a proteomic signature using ML-based MS. The test evaluates the spectrum of blood samples and assigns patients to either VeriStrat "good" (VS-G) or VeriStrat "poor" (VS-P) groups. It was found that the VeriStrat status was significantly associated with both PFS and OS in NSCLC patients receiving ICI treatment [80].
Compared to genomics and transcriptomics, proteomics may be more effective in assessing features of the TME and immune responses in patients with lung cancer, resulting in a more accurate prediction of the response to immunotherapy. The application of AI methods to proteomics may thus represent a future direction in the development of improved methods for assessing patient prognosis after immunotherapy.

AI-based microbiomics

Many factors associated with the gut microbiota, including its composition and community structures, influence the immunotherapy response. Thus, statistical analysis using AI can assist in the elucidation of the specific composition or important combinations of gut microbiota species. Tatsuro Okamoto will conduct a prospective observational study using AI (UMIN000046428) to elucidate the specific gut microbiome composition or gut microbial combinations associated with the immunotherapy response in lung cancer patients [81].

AI-based radiomics

Based on contrast-enhanced CT before neoadjuvant immunotherapy, a study used radiomic features, clinicopathological information, and DL features to construct a model for the prediction of a good pathological response, finding an AUC of 0.805 [82]. In another study, a combination of radiomics and DL was used for predicting the response of NSCLC patients with advanced disease to immunotherapy with an AUC of 0.960 [83]. He et al. also demonstrated that the combination of radiomics with DL could identify patients likely to benefit from ICI treatment (p < 0.001) [84].
Radiomics is both non-invasive and reproducible and thus offers a novel solution. Also, radiomics-based predictive models allow the early identification of suitable patients for immunotherapy, thus enabling precision medicine.

Future challenges

Although various studies have explored the potential of various histological approaches and AI in predicting the response to immunotherapy, challenges associated with their application in clinical practice still remain.
Both the methods of measurement and the platforms used to measure most genomic biomarkers are inconsistent, resulting in poor reliability and reduced accuracy. Furthermore, the optimal threshold is difficult to determine, i.e., different studies have used different thresholds for prediction, leading to inconsistent results. MS-based proteomics has been widely used in cancer immunology research, but MS proteomics has difficulty in accurately identifying unique peptides and different protein isoforms. Transcriptomics also presents several difficulties, specifically, in relation to sample preparation, computational analysis, and reproducibility. As for microbiomics, its future challenges include an inadequate understanding of the modulating effects of the microbiota on the therapeutic response, a lack of information and consensus on the use of microbial signatures as predictive biomarkers, and limited information on how the microbiota could be modulated. In addition, while current research has focused on bacteria, symbiotic viruses, fungi, and archaea also have non-negligible roles in cancer. In terms of radiomics, a significant limitation is the absence of standardization between studies as this complicates data sharing and reduces the generalizability of models constructed in different institutions (Table 1).
Table 1
Advantages and disadvantages of biomarkers for predicting immunotherapy outcome
Category
Advantage
Disadvantages
PD-L1
1. Easy to detect
2. FDA-approved
1. Inconsistent detection methods
2. Uncertain optimal cutoff value
3. PD-L1 dynamic changes
TMB
1. FDA-approved
2. Independent of other predictors such as PD-L1 expression
1. Tumor spatiotemporal heterogeneity
2. Inconsistent detection methods
3. Uncertain optimal cutoff value
ctDNA
1. Minimally invasive
2. High repeatability
1.Detection technology and standardization issues
2. ctDNA source and decomposition issues
NAL
1. Improve the accuracy of treatment
1. Technical limitations
2. High data quality requirements
3. Difficulty obtaining data
Gene mutation
1. Wide clinical application
1. Inconsistent detection platform and analysis method
2. Uncertainty in predictions
3. Tumor heterogeneity and dynamics
Transcriptome
1. Aid clinical decision-making
2. Overall
1. Difficult data analysis
2. Uncertainty about data sources
3. Lack of uniform standards
Proteome
1. Minimally invasive (usually only plasma)
1. Protein marker diversity
2. Technical complexity
3. Difficulty in interpreting data
Microbiome
1. Easy access to samples
2. High repeatability
1. Lack of standardized methods
2. Unclear mechanism
Radiomics
1. Non-invasive
2.Multi-parameter analysis
3. High precision
4. Fast
1. Sample size limitation
2. Difficult data analysis
3. High cost
Faced with the challenge of lack of standardization and validation of biomarkers for predicting the efficacy of immunotherapy in NSCLC, the following are some suggestions and directions for future efforts: 1) Standardize measurement methods of biomarkers: Develop unified measurement methods and technical standards to ensure the reliability and comparability of biomarker measurement results. Establish standard operating procedures and quality control procedures to reduce measurement errors. 2) Multi-center collaborative research: Establish an international or cross-institutional research collaboration organization to conduct large-scale research, collect more sample data, and conduct verification and comparative analysis of different immunotherapy drugs and treatment options. Cross-center collaboration can increase the number of samples and improve the credibility of the study. 3) Unified data collection and sharing: Establish a unified database for collecting and storing biomarker data generated in clinical trials and real-world applications. By sharing data, the verification and promotion of biomarkers can be accelerated, and more data can be provided for the discovery and verification of future biomarkers.
The cost-effectiveness of different biomarkers also deserves discussion. Because genomics and radiomics can use existing gene sequencing data and existing medical imaging data, their application in efficacy prediction is relatively low-cost. Microbiomics is less expensive but requires steps such as sample collection and high-throughput sequencing. Transcriptomics and proteomics involve multiple steps such as sample collection, RNA (protein) extraction, sequencing, and analysis, so their costs are relatively high. In low-resource settings, several strategies can help improve the cost-effectiveness: 1) Optimize sample collection and processing procedures to ensure high-quality data acquisition. 2) Reasonably use existing data resources and conduct secondary analysis to reduce the cost of new data collection. 3) Establish cross-institutional or international collaboration networks to share data and resources. 4) Develop simpler, cost-effective measurement technologies, such as rapid sequencing, portable imaging equipment, etc. 5) Further strengthen the development of artificial intelligence algorithms and improve the accuracy and efficiency of prediction models, thereby saving the cost of data analysis and interpretation.

Conclusions

In this study, we reviewed the application of genomics, transcriptomics, proteomics, microbiomics, and radiomics in predicting immunotherapy outcomes. While data on the use of individual biomarkers to predict response to immunotherapy are plentiful, direct comparisons between them are scarce. PD-L1 is still the most commonly used, and its combination with other biomarkers can help improve the predictive ability of immunotherapy efficacy. In addition, the combination of AI and multi-omics data can help integrate the various data and realize automatic prediction, ultimately providing personalized treatment for patients with lung cancer.

Acknowledgements

This project was supported by a grant from the Regional Innovation Cooperation Project of the Sichuan Science and Technology Program (2021YFQ0029).

Declarations

Competing interests

The authors have no relevant financial or non-financial interests to disclose.

Ethics approval

Not applicable.
Not applicable.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Jetzt bestellen und 100 € sparen!

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Jetzt bestellen und 100 € sparen!

Literatur
1.
Zurück zum Zitat Siegel RL, et al. Cancer Statistics, 2021. CA: a Cancer J Clin. 2021;71(1):7-33. Siegel RL, et al. Cancer Statistics, 2021. CA: a Cancer J Clin. 2021;71(1):7-33.
2.
Zurück zum Zitat Reck M, et al. Management of non-small-cell lung cancer: recent developments. Lancet (London, England). 2013;382(9893):709–19.PubMedCrossRef Reck M, et al. Management of non-small-cell lung cancer: recent developments. Lancet (London, England). 2013;382(9893):709–19.PubMedCrossRef
3.
Zurück zum Zitat Park K, et al. Pan-Asian adapted ESMO clinical practice guidelines for the management of patients with locally-advanced unresectable non-small-cell lung cancer: a KSMO-ESMO initiative endorsed by CSCO, ISMPO, JSMO, MOS SSO and TOS. Ann Oncol. 2020;31(2):191–201.PubMedCrossRef Park K, et al. Pan-Asian adapted ESMO clinical practice guidelines for the management of patients with locally-advanced unresectable non-small-cell lung cancer: a KSMO-ESMO initiative endorsed by CSCO, ISMPO, JSMO, MOS SSO and TOS. Ann Oncol. 2020;31(2):191–201.PubMedCrossRef
4.
Zurück zum Zitat Rittmeyer A, et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. The Lancet. 2017;389(10066):255–65.CrossRef Rittmeyer A, et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. The Lancet. 2017;389(10066):255–65.CrossRef
5.
Zurück zum Zitat Suresh K, et al. Immune checkpoint immunotherapy for non-small cell lung cancer: benefits and pulmonary toxicities. Chest. 2018;154(6):1416–23.PubMedPubMedCentralCrossRef Suresh K, et al. Immune checkpoint immunotherapy for non-small cell lung cancer: benefits and pulmonary toxicities. Chest. 2018;154(6):1416–23.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Ettinger DS, et al. NCCN guidelines insights: non-small cell lung cancer. J Natl Compr Cancer Netw: JNCCN. 2021;19(3):254–66.PubMedCrossRef Ettinger DS, et al. NCCN guidelines insights: non-small cell lung cancer. J Natl Compr Cancer Netw: JNCCN. 2021;19(3):254–66.PubMedCrossRef
9.
11.
Zurück zum Zitat Jia Q, et al. Tracking neoantigens by personalized circulating tumor DNA sequencing during checkpoint blockade immunotherapy in non-small cell lung cancer. Adv Sci (Weinh). 2020;7(9):1903410.PubMedCrossRef Jia Q, et al. Tracking neoantigens by personalized circulating tumor DNA sequencing during checkpoint blockade immunotherapy in non-small cell lung cancer. Adv Sci (Weinh). 2020;7(9):1903410.PubMedCrossRef
12.
Zurück zum Zitat Kantarjian H, Yu PP. Artificial intelligence, big data, and cancer. JAMA Oncol. 2015;1(5):573–4.PubMedCrossRef Kantarjian H, Yu PP. Artificial intelligence, big data, and cancer. JAMA Oncol. 2015;1(5):573–4.PubMedCrossRef
14.
Zurück zum Zitat Yan X, Ding J, Cheng HD. A Novel Adaptive Fuzzy Deep Learning Approach for Histopathologic Cancer Detection. Annu Int Conf IEEE Eng Med Biol Soc. 2021 Nov;2021:3518–3521. Yan X, Ding J, Cheng HD. A Novel Adaptive Fuzzy Deep Learning Approach for Histopathologic Cancer Detection. Annu Int Conf IEEE Eng Med Biol Soc. 2021 Nov;2021:3518–3521.
15.
Zurück zum Zitat Serag A, et al. Translational AI and deep learning in diagnostic pathology. Front Med. 2019;6:185.CrossRef Serag A, et al. Translational AI and deep learning in diagnostic pathology. Front Med. 2019;6:185.CrossRef
16.
Zurück zum Zitat Dong Y, et al. Multi-channel multi-task deep learning for predicting EGFR and KRAS mutations of non-small cell lung cancer on CT images. Quant Imaging Med Surg. 2021;11(6):2354–75.PubMedPubMedCentralCrossRef Dong Y, et al. Multi-channel multi-task deep learning for predicting EGFR and KRAS mutations of non-small cell lung cancer on CT images. Quant Imaging Med Surg. 2021;11(6):2354–75.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Davoli T, et al. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science. 2017;355(6322):eaaf8399.PubMedPubMedCentralCrossRef Davoli T, et al. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science. 2017;355(6322):eaaf8399.PubMedPubMedCentralCrossRef
19.
20.
Zurück zum Zitat Peters S, et al. Atezolizumab versus chemotherapy in advanced or metastatic NSCLC with high blood-based tumor mutational burden: primary analysis of BFAST cohort C randomized phase 3 trial. Nat Med. 2022;28(9):1831–9.PubMedPubMedCentralCrossRef Peters S, et al. Atezolizumab versus chemotherapy in advanced or metastatic NSCLC with high blood-based tumor mutational burden: primary analysis of BFAST cohort C randomized phase 3 trial. Nat Med. 2022;28(9):1831–9.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Singal G, et al. Association of patient characteristics and tumor genomics with clinical outcomes among patients with non-small cell lung cancer using a clinicogenomic database. JAMA. 2019;321(14):1391–9.PubMedPubMedCentralCrossRef Singal G, et al. Association of patient characteristics and tumor genomics with clinical outcomes among patients with non-small cell lung cancer using a clinicogenomic database. JAMA. 2019;321(14):1391–9.PubMedPubMedCentralCrossRef
22.
Zurück zum Zitat Stover DG, et al. Association of cell-free DNA tumor fraction and somatic copy number alterations with survival in metastatic triple-negative breast cancer. J Clin Oncol. 2018;36(6):543–53.PubMedPubMedCentralCrossRef Stover DG, et al. Association of cell-free DNA tumor fraction and somatic copy number alterations with survival in metastatic triple-negative breast cancer. J Clin Oncol. 2018;36(6):543–53.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Bernard V, et al. Circulating nucleic acids are associated with outcomes of patients with pancreatic cancer. Gastroenterology. 2019;156(1):108-118.e4.PubMedCrossRef Bernard V, et al. Circulating nucleic acids are associated with outcomes of patients with pancreatic cancer. Gastroenterology. 2019;156(1):108-118.e4.PubMedCrossRef
24.
Zurück zum Zitat Bratman SV, et al. Personalized circulating tumor DNA analysis as a predictive biomarker in solid tumor patients treated with pembrolizumab. Nature Cancer. 2020;1(9):873–81.PubMedCrossRef Bratman SV, et al. Personalized circulating tumor DNA analysis as a predictive biomarker in solid tumor patients treated with pembrolizumab. Nature Cancer. 2020;1(9):873–81.PubMedCrossRef
25.
Zurück zum Zitat Ricciuti B, et al. Early plasma circulating tumor DNA (ctDNA) changes predict response to first-line pembrolizumab-based therapy in non-small cell lung cancer (NSCLC). J Immunother Cancer. 2021;9(3): e001504.PubMedPubMedCentralCrossRef Ricciuti B, et al. Early plasma circulating tumor DNA (ctDNA) changes predict response to first-line pembrolizumab-based therapy in non-small cell lung cancer (NSCLC). J Immunother Cancer. 2021;9(3): e001504.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Vokes NI, et al. Circulating tumor DNA (ctDNA) dynamics and survival outcomes in patients (pts) with advanced non-small cell lung cancer (aNSCLC) and high (>50%) programmed cell death-ligand 1 (PD-L1) expression, randomized to cemiplimab (cemi) vs chemotherapy (chemo). J Clin Oncol. 2023;41(16_suppl):9022–9022.CrossRef Vokes NI, et al. Circulating tumor DNA (ctDNA) dynamics and survival outcomes in patients (pts) with advanced non-small cell lung cancer (aNSCLC) and high (>50%) programmed cell death-ligand 1 (PD-L1) expression, randomized to cemiplimab (cemi) vs chemotherapy (chemo). J Clin Oncol. 2023;41(16_suppl):9022–9022.CrossRef
27.
Zurück zum Zitat Guibert N, et al. Monitoring of KRAS -mutated ctDNA to discriminate pseudo-progression from true progression during anti-PD-1 treatment of lung adenocarcinoma. Oncotarget. 2017;8(23):38056.PubMedPubMedCentralCrossRef Guibert N, et al. Monitoring of KRAS -mutated ctDNA to discriminate pseudo-progression from true progression during anti-PD-1 treatment of lung adenocarcinoma. Oncotarget. 2017;8(23):38056.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Lee CK, et al. Checkpoint inhibitors in metastatic EGFR-mutated non–small cell lung cancer—a meta-analysis. J Thorac Oncol. 2017;12(2):403–7.PubMedCrossRef Lee CK, et al. Checkpoint inhibitors in metastatic EGFR-mutated non–small cell lung cancer—a meta-analysis. J Thorac Oncol. 2017;12(2):403–7.PubMedCrossRef
29.
Zurück zum Zitat Lee CK, et al. Clinical and molecular characteristics associated with survival among patients treated with checkpoint inhibitors for advanced non-small cell lung carcinoma: a systematic review and meta-analysis. JAMA Oncol. 2018;4(2):210–6.PubMedCrossRef Lee CK, et al. Clinical and molecular characteristics associated with survival among patients treated with checkpoint inhibitors for advanced non-small cell lung carcinoma: a systematic review and meta-analysis. JAMA Oncol. 2018;4(2):210–6.PubMedCrossRef
30.
Zurück zum Zitat Rizvi NA, et al. Nivolumab in combination with platinum-based doublet chemotherapy for first-line treatment of advanced non-small-cell lung cancer. J Clin Oncol. 2016;34(25):2969–79.PubMedPubMedCentralCrossRef Rizvi NA, et al. Nivolumab in combination with platinum-based doublet chemotherapy for first-line treatment of advanced non-small-cell lung cancer. J Clin Oncol. 2016;34(25):2969–79.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Lu S, et al. VP9-2021: ORIENT-31: Phase III study of sintilimab with or without IBI305 plus chemotherapy in patients with EGFR mutated nonsquamous NSCLC who progressed after EGFR-TKI therapy. Ann Oncol. 2022;33(1):112–3.CrossRef Lu S, et al. VP9-2021: ORIENT-31: Phase III study of sintilimab with or without IBI305 plus chemotherapy in patients with EGFR mutated nonsquamous NSCLC who progressed after EGFR-TKI therapy. Ann Oncol. 2022;33(1):112–3.CrossRef
32.
Zurück zum Zitat Socinski MA, et al. IMpower150 final overall survival analyses for atezolizumab plus bevacizumab and chemotherapy in first-line metastatic nonsquamous NSCLC. J Thorac Oncol. 2021;16(11):1909–24.PubMedCrossRef Socinski MA, et al. IMpower150 final overall survival analyses for atezolizumab plus bevacizumab and chemotherapy in first-line metastatic nonsquamous NSCLC. J Thorac Oncol. 2021;16(11):1909–24.PubMedCrossRef
33.
Zurück zum Zitat Park S, et al. Phase III, Randomized Study of Atezolizumab Plus Bevacizumab and Chemotherapy in Patients With EGFR- or ALK-Mutated Non-Small-Cell Lung Cancer (ATTLAS, KCSG-LU19-04). J Clin Oncol. 2023 Oct 20:JCO2301891. Park S, et al. Phase III, Randomized Study of Atezolizumab Plus Bevacizumab and Chemotherapy in Patients With EGFR- or ALK-Mutated Non-Small-Cell Lung Cancer (ATTLAS, KCSG-LU19-04). J Clin Oncol. 2023 Oct 20:JCO2301891.
34.
Zurück zum Zitat Mok T, et al. Nivolumab Plus Chemotherapy in Epidermal Growth Factor Receptor-Mutated Metastatic Non-Small-Cell Lung Cancer After Disease Progression on Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors: Final Results of CheckMate 722. J Clin Oncol. 2024 Jan 22:JCO2301017. Mok T, et al. Nivolumab Plus Chemotherapy in Epidermal Growth Factor Receptor-Mutated Metastatic Non-Small-Cell Lung Cancer After Disease Progression on Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors: Final Results of CheckMate 722. J Clin Oncol. 2024 Jan 22:JCO2301017.
35.
Zurück zum Zitat Yang JC-H, et al. Pemetrexed and platinum with or without pembrolizumab for tyrosine kinase inhibitor (TKI)-resistant, EGFR-mutant, metastatic nonsquamous NSCLC: Phase 3 KEYNOTE-789 study. J Clin Oncol. 2023;41(17_suppl):9000.CrossRef Yang JC-H, et al. Pemetrexed and platinum with or without pembrolizumab for tyrosine kinase inhibitor (TKI)-resistant, EGFR-mutant, metastatic nonsquamous NSCLC: Phase 3 KEYNOTE-789 study. J Clin Oncol. 2023;41(17_suppl):9000.CrossRef
36.
Zurück zum Zitat Negrao MV, et al. Oncogene-specific differences in tumor mutational burden, PD-L1 expression, and outcomes from immunotherapy in non-small cell lung cancer. J immunother cancer. 2021;9(8):e002891.PubMedPubMedCentralCrossRef Negrao MV, et al. Oncogene-specific differences in tumor mutational burden, PD-L1 expression, and outcomes from immunotherapy in non-small cell lung cancer. J immunother cancer. 2021;9(8):e002891.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Garassino MC, et al. Durvalumab as third-line or later treatment for advanced non-small-cell lung cancer (ATLANTIC): an open-label, single-arm, phase 2 study. Lancet Oncol. 2018;19(4):521–36.PubMedPubMedCentralCrossRef Garassino MC, et al. Durvalumab as third-line or later treatment for advanced non-small-cell lung cancer (ATLANTIC): an open-label, single-arm, phase 2 study. Lancet Oncol. 2018;19(4):521–36.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Mazieres J, et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: results from the IMMUNOTARGET registry. Ann Oncol. 2019;30(8):1321–8.PubMedPubMedCentralCrossRef Mazieres J, et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: results from the IMMUNOTARGET registry. Ann Oncol. 2019;30(8):1321–8.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Offin M, et al. Immunophenotype and response to immunotherapy of RET-rearranged lung cancers. JCO Precis Oncol. 2019;3:1–8.CrossRef Offin M, et al. Immunophenotype and response to immunotherapy of RET-rearranged lung cancers. JCO Precis Oncol. 2019;3:1–8.CrossRef
40.
Zurück zum Zitat Lee J, et al. Characteristics and outcomes of RET-rearranged Korean non-small cell lung cancer patients in real-world practice. Jpn J Clin Oncol. 2020;50(5):594–601.PubMedCrossRef Lee J, et al. Characteristics and outcomes of RET-rearranged Korean non-small cell lung cancer patients in real-world practice. Jpn J Clin Oncol. 2020;50(5):594–601.PubMedCrossRef
41.
42.
Zurück zum Zitat Yang G, et al. First-line immunotherapy or angiogenesis inhibitor plus chemotherapy for HER2-altered NSCLC: a retrospective real-world POLISH study. Ther Adv Med Oncol. 2022;14:17588359221082340.PubMedPubMedCentralCrossRef Yang G, et al. First-line immunotherapy or angiogenesis inhibitor plus chemotherapy for HER2-altered NSCLC: a retrospective real-world POLISH study. Ther Adv Med Oncol. 2022;14:17588359221082340.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Calles A, et al. Expression of PD-1 and its ligands, PD-L1 and PD-L2, in smokers and never smokers with KRAS-mutant lung cancer. J Thorac Oncol. 2015;10(12):1726–35.PubMedCrossRef Calles A, et al. Expression of PD-1 and its ligands, PD-L1 and PD-L2, in smokers and never smokers with KRAS-mutant lung cancer. J Thorac Oncol. 2015;10(12):1726–35.PubMedCrossRef
44.
Zurück zum Zitat Erica C. Nakajima, et al. Outcomes of first-line immune checkpoint inhibitors with or without chemotherapy according to KRAS mutational status and PD-L1 expression in patients with advanced NSCLC: FDA pooled analysis.. JCO 40, 9001–9001(2022). Erica C. Nakajima, et al. Outcomes of first-line immune checkpoint inhibitors with or without chemotherapy according to KRAS mutational status and PD-L1 expression in patients with advanced NSCLC: FDA pooled analysis.. JCO 40, 9001–9001(2022).
45.
Zurück zum Zitat Noordhof A, et al. Prognostic impact of KRAS mutation status for patients with stage IV adenocarcinoma of the lung treated with first-line pembrolizumab monotherapy. Lung Cancer. 2021;155:163–9.PubMedCrossRef Noordhof A, et al. Prognostic impact of KRAS mutation status for patients with stage IV adenocarcinoma of the lung treated with first-line pembrolizumab monotherapy. Lung Cancer. 2021;155:163–9.PubMedCrossRef
46.
Zurück zum Zitat Ricciuti B, et al. Dissecting the clinicopathologic, genomic, and immunophenotypic correlates of KRAS(G12D)-mutated non-small-cell lung cancer. Ann Oncol. 2022;33(10):1029–40.PubMedCrossRef Ricciuti B, et al. Dissecting the clinicopathologic, genomic, and immunophenotypic correlates of KRAS(G12D)-mutated non-small-cell lung cancer. Ann Oncol. 2022;33(10):1029–40.PubMedCrossRef
47.
48.
Zurück zum Zitat Dong Z-Y, et al. Potential predictive value of TP53 and KRAS mutation status for response to PD-1 blockade immunotherapy in lung adenocarcinoma. Clin Cancer Res. 2017;23(12):3012–24.PubMedCrossRef Dong Z-Y, et al. Potential predictive value of TP53 and KRAS mutation status for response to PD-1 blockade immunotherapy in lung adenocarcinoma. Clin Cancer Res. 2017;23(12):3012–24.PubMedCrossRef
49.
Zurück zum Zitat Chen X, et al. Pan-cancer analysis of KEAP1 mutations as biomarkers for immunotherapy outcomes. Ann Transl Med. 2019;8(4):141.CrossRef Chen X, et al. Pan-cancer analysis of KEAP1 mutations as biomarkers for immunotherapy outcomes. Ann Transl Med. 2019;8(4):141.CrossRef
50.
Zurück zum Zitat Alessi JV, et al. SMARCA4 and other SWItch/Sucrose nonfermentable family genomic alterations in NSCLC: clinicopathologic characteristics and outcomes to immune checkpoint inhibition. J Thorac Oncol. 2021;16(7):1176–87.PubMedCrossRef Alessi JV, et al. SMARCA4 and other SWItch/Sucrose nonfermentable family genomic alterations in NSCLC: clinicopathologic characteristics and outcomes to immune checkpoint inhibition. J Thorac Oncol. 2021;16(7):1176–87.PubMedCrossRef
51.
Zurück zum Zitat Murciano-Goroff YR, et al. Immune biomarkers and response to checkpoint inhibition of BRAF V600 and BRAF non-V600 altered lung cancers. Br J Cancer. 2022;126(6):889–98.PubMedCrossRef Murciano-Goroff YR, et al. Immune biomarkers and response to checkpoint inhibition of BRAF V600 and BRAF non-V600 altered lung cancers. Br J Cancer. 2022;126(6):889–98.PubMedCrossRef
52.
Zurück zum Zitat Li H, et al. Tumor immune microenvironment and immunotherapy efficacy in BRAF mutation non-small-cell lung cancer. Cell Death Dis. 2022;13(12):1064.PubMedPubMedCentralCrossRef Li H, et al. Tumor immune microenvironment and immunotherapy efficacy in BRAF mutation non-small-cell lung cancer. Cell Death Dis. 2022;13(12):1064.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Guisier F, et al. Efficacy and safety of Anti–PD-1 immunotherapy in patients with advanced NSCLC With BRAF, HER2, or MET mutations or RET translocation: GFPC 01–2018. J Thorac Oncol. 2020;15(4):628–36.PubMedCrossRef Guisier F, et al. Efficacy and safety of Anti–PD-1 immunotherapy in patients with advanced NSCLC With BRAF, HER2, or MET mutations or RET translocation: GFPC 01–2018. J Thorac Oncol. 2020;15(4):628–36.PubMedCrossRef
54.
Zurück zum Zitat Sabari JK, et al. PD-L1 expression, tumor mutational burden, and response to immunotherapy in patients with MET exon 14 altered lung cancers. Ann Oncol. 2018;29(10):2085–91.PubMedPubMedCentralCrossRef Sabari JK, et al. PD-L1 expression, tumor mutational burden, and response to immunotherapy in patients with MET exon 14 altered lung cancers. Ann Oncol. 2018;29(10):2085–91.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Cieślik M, Chinnaiyan AM. Cancer transcriptome profiling at the juncture of clinical translation. Nat Rev Genet. 2018;19(2):93–109.PubMedCrossRef Cieślik M, Chinnaiyan AM. Cancer transcriptome profiling at the juncture of clinical translation. Nat Rev Genet. 2018;19(2):93–109.PubMedCrossRef
57.
Zurück zum Zitat Ott PA, et al. T-cell-inflamed gene-expression profile, programmed death ligand 1 expression, and tumor mutational burden predict efficacy in patients treated with pembrolizumab across 20 cancers: KEYNOTE-028. J Clin Oncol. 2019;37(4):318–27.PubMedCrossRef Ott PA, et al. T-cell-inflamed gene-expression profile, programmed death ligand 1 expression, and tumor mutational burden predict efficacy in patients treated with pembrolizumab across 20 cancers: KEYNOTE-028. J Clin Oncol. 2019;37(4):318–27.PubMedCrossRef
58.
Zurück zum Zitat Fehrenbacher L, et al. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial. Lancet. 2016;387(10030):1837–46.PubMedCrossRef Fehrenbacher L, et al. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial. Lancet. 2016;387(10030):1837–46.PubMedCrossRef
59.
Zurück zum Zitat Higgs BW, et al. Interferon gamma messenger RNA signature in tumor biopsies predicts outcomes in patients with non-small cell lung carcinoma or urothelial cancer treated with durvalumab. Clin Cancer Res. 2018;24(16):3857–66.PubMedCrossRef Higgs BW, et al. Interferon gamma messenger RNA signature in tumor biopsies predicts outcomes in patients with non-small cell lung carcinoma or urothelial cancer treated with durvalumab. Clin Cancer Res. 2018;24(16):3857–66.PubMedCrossRef
60.
Zurück zum Zitat Moutafi MK, et al. Spatially resolved proteomic profiling identifies tumor cell CD44 as a biomarker associated with sensitivity to PD-1 axis blockade in advanced non-small-cell lung cancer. J Immunother Cancer. 2022;10(8):e004757.PubMedPubMedCentralCrossRef Moutafi MK, et al. Spatially resolved proteomic profiling identifies tumor cell CD44 as a biomarker associated with sensitivity to PD-1 axis blockade in advanced non-small-cell lung cancer. J Immunother Cancer. 2022;10(8):e004757.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Shen CY, et al. Tissue proteogenomic landscape reveals the role of uncharacterized SEL1L3 in progression and immunotherapy response in lung adenocarcinoma. J Proteome Res. 2022;22(4):1056–70.PubMedCrossRef Shen CY, et al. Tissue proteogenomic landscape reveals the role of uncharacterized SEL1L3 in progression and immunotherapy response in lung adenocarcinoma. J Proteome Res. 2022;22(4):1056–70.PubMedCrossRef
62.
Zurück zum Zitat Rich P, et al. Real-world performance of blood-based proteomic profiling in first-line immunotherapy treatment in advanced stage non-small cell lung cancer. J Immunother Cancer. 2021;9(10):e002989.PubMedPubMedCentralCrossRef Rich P, et al. Real-world performance of blood-based proteomic profiling in first-line immunotherapy treatment in advanced stage non-small cell lung cancer. J Immunother Cancer. 2021;9(10):e002989.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Eltahir M, et al. Plasma proteomic analysis in non-small cell lung cancer patients treated with PD-1/PD-L1 blockade. Cancers (Basel). 2021;13(13):3116.PubMedCrossRef Eltahir M, et al. Plasma proteomic analysis in non-small cell lung cancer patients treated with PD-1/PD-L1 blockade. Cancers (Basel). 2021;13(13):3116.PubMedCrossRef
64.
Zurück zum Zitat Routy B, et al. Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–7.PubMedCrossRef Routy B, et al. Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–7.PubMedCrossRef
65.
Zurück zum Zitat Derosa L, et al. Intestinal Akkermansia muciniphila predicts overall survival in advanced non-small cell lung cancer patients treated with anti-PD-1 antibodies: results a phase II study. J Clin Oncol. 2021;39(15_suppl):9019–9019.CrossRef Derosa L, et al. Intestinal Akkermansia muciniphila predicts overall survival in advanced non-small cell lung cancer patients treated with anti-PD-1 antibodies: results a phase II study. J Clin Oncol. 2021;39(15_suppl):9019–9019.CrossRef
66.
Zurück zum Zitat Boesch M, et al. Local tumor microbial signatures and response to checkpoint blockade in non-small cell lung cancer. Oncoimmunology. 2021;10(1):1988403.PubMedPubMedCentralCrossRef Boesch M, et al. Local tumor microbial signatures and response to checkpoint blockade in non-small cell lung cancer. Oncoimmunology. 2021;10(1):1988403.PubMedPubMedCentralCrossRef
67.
68.
Zurück zum Zitat Tunali I, et al. P101–041 quantitative imaging features predict response of immunotherapy in non-small cell lung cancer patients: topic: prognostic factors Treatment. J Thorac Oncol. 2017;12(1):S474–5.CrossRef Tunali I, et al. P101–041 quantitative imaging features predict response of immunotherapy in non-small cell lung cancer patients: topic: prognostic factors Treatment. J Thorac Oncol. 2017;12(1):S474–5.CrossRef
69.
Zurück zum Zitat Yang B, et al. Combination of computed tomography imaging-based radiomics and clinicopathological characteristics for predicting the clinical benefits of immune checkpoint inhibitors in lung cancer. Respir Res. 2021;22(1):189.PubMedPubMedCentralCrossRef Yang B, et al. Combination of computed tomography imaging-based radiomics and clinicopathological characteristics for predicting the clinical benefits of immune checkpoint inhibitors in lung cancer. Respir Res. 2021;22(1):189.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Sun R, et al. A radiomics approach to assess tumour-infiltrating CD8 cells and response to anti-PD-1 or anti-PD-L1 immunotherapy: an imaging biomarker, retrospective multicohort study. Lancet Oncol. 2018;19(9):1180–91.PubMedCrossRef Sun R, et al. A radiomics approach to assess tumour-infiltrating CD8 cells and response to anti-PD-1 or anti-PD-L1 immunotherapy: an imaging biomarker, retrospective multicohort study. Lancet Oncol. 2018;19(9):1180–91.PubMedCrossRef
71.
Zurück zum Zitat Vaidya P, et al. Novel, non-invasive imaging approach to identify patients with advanced non-small cell lung cancer at risk of hyperprogressive disease with immune checkpoint blockade. J Immunother Cancer. 2020;8(2):e001343.PubMedPubMedCentralCrossRef Vaidya P, et al. Novel, non-invasive imaging approach to identify patients with advanced non-small cell lung cancer at risk of hyperprogressive disease with immune checkpoint blockade. J Immunother Cancer. 2020;8(2):e001343.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Tunali I, et al. Novel clinical and radiomic predictors of rapid disease progression phenotypes among lung cancer patients treated with immunotherapy: an early report. Lung Cancer. 2019;129:75–9.PubMedCrossRef Tunali I, et al. Novel clinical and radiomic predictors of rapid disease progression phenotypes among lung cancer patients treated with immunotherapy: an early report. Lung Cancer. 2019;129:75–9.PubMedCrossRef
73.
Zurück zum Zitat Barabino E, et al. Exploring response to immunotherapy in non-small cell lung cancer using delta-radiomics. Cancers (Basel). 2022;14(2):350.PubMedCrossRef Barabino E, et al. Exploring response to immunotherapy in non-small cell lung cancer using delta-radiomics. Cancers (Basel). 2022;14(2):350.PubMedCrossRef
74.
Zurück zum Zitat He B, et al. Predicting response to immunotherapy in advanced non-small-cell lung cancer using tumor mutational burden radiomic biomarker. J Immunother Cancer. 2020;8(2):e000550.PubMedPubMedCentralCrossRef He B, et al. Predicting response to immunotherapy in advanced non-small-cell lung cancer using tumor mutational burden radiomic biomarker. J Immunother Cancer. 2020;8(2):e000550.PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Jain MS, Massoud TF. Predicting tumour mutational burden from histopathological images using multiscale deep learning. Nat Mach Intell. 2020;2(6):356–62.CrossRef Jain MS, Massoud TF. Predicting tumour mutational burden from histopathological images using multiscale deep learning. Nat Mach Intell. 2020;2(6):356–62.CrossRef
76.
Zurück zum Zitat Suphavilai C, et al. Predicting heterogeneity in clone-specific therapeutic vulnerabilities using single-cell transcriptomic signatures. Genome Med. 2021;13(1):189.PubMedPubMedCentralCrossRef Suphavilai C, et al. Predicting heterogeneity in clone-specific therapeutic vulnerabilities using single-cell transcriptomic signatures. Genome Med. 2021;13(1):189.PubMedPubMedCentralCrossRef
77.
78.
79.
Zurück zum Zitat Muller M, et al. A serum protein classifier identifying patients with advanced non-small cell lung cancer who derive clinical benefit from treatment with immune checkpoint inhibitors. Clin Cancer Res. 2020;26(19):5188–97.PubMedCrossRef Muller M, et al. A serum protein classifier identifying patients with advanced non-small cell lung cancer who derive clinical benefit from treatment with immune checkpoint inhibitors. Clin Cancer Res. 2020;26(19):5188–97.PubMedCrossRef
80.
Zurück zum Zitat Chae YK, et al. Mass spectrometry-based serum proteomic signature as a potential biomarker for survival in patients with non-small cell lung cancer receiving immunotherapy. Transl Lung Cancer Res. 2020;9(4):1015–28.PubMedPubMedCentralCrossRef Chae YK, et al. Mass spectrometry-based serum proteomic signature as a potential biomarker for survival in patients with non-small cell lung cancer receiving immunotherapy. Transl Lung Cancer Res. 2020;9(4):1015–28.PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Shoji F, et al. Artificial intelligence-derived gut microbiome as a predictive biomarker for therapeutic response to immunotherapy in lung cancer: protocol for a multicentre, prospective, observational study. BMJ Open. 2022;12(6): e061674.PubMedPubMedCentralCrossRef Shoji F, et al. Artificial intelligence-derived gut microbiome as a predictive biomarker for therapeutic response to immunotherapy in lung cancer: protocol for a multicentre, prospective, observational study. BMJ Open. 2022;12(6): e061674.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Lin Q, et al. CT-based radiomics in predicting pathological response in non-small cell lung cancer patients receiving neoadjuvant immunotherapy. Front Oncol. 2022;12: 937277.PubMedPubMedCentralCrossRef Lin Q, et al. CT-based radiomics in predicting pathological response in non-small cell lung cancer patients receiving neoadjuvant immunotherapy. Front Oncol. 2022;12: 937277.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Ren Q, et al. Assessing the robustness of radiomics/deep learning approach in the identification of efficacy of anti-PD-1 treatment in advanced or metastatic non-small cell lung carcinoma patients. Front Oncol. 2022;12: 952749.PubMedPubMedCentralCrossRef Ren Q, et al. Assessing the robustness of radiomics/deep learning approach in the identification of efficacy of anti-PD-1 treatment in advanced or metastatic non-small cell lung carcinoma patients. Front Oncol. 2022;12: 952749.PubMedPubMedCentralCrossRef
84.
Zurück zum Zitat He BX, et al. Deep learning for predicting immunotherapeutic efficacy in advanced non-small cell lung cancer patients: a retrospective study combining progression-free survival risk and overall survival risk. Transl Lung Cancer Res. 2022;11(4):670–85.PubMedPubMedCentralCrossRef He BX, et al. Deep learning for predicting immunotherapeutic efficacy in advanced non-small cell lung cancer patients: a retrospective study combining progression-free survival risk and overall survival risk. Transl Lung Cancer Res. 2022;11(4):670–85.PubMedPubMedCentralCrossRef
Metadaten
Titel
Multi-omics and artificial intelligence predict clinical outcomes of immunotherapy in non-small cell lung cancer patients
verfasst von
Ting Mei
Ting Wang
Qinghua Zhou
Publikationsdatum
01.12.2024
Verlag
Springer International Publishing
Erschienen in
Clinical and Experimental Medicine / Ausgabe 1/2024
Print ISSN: 1591-8890
Elektronische ISSN: 1591-9528
DOI
https://doi.org/10.1007/s10238-024-01324-0

Weitere Artikel der Ausgabe 1/2024

Clinical and Experimental Medicine 1/2024 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.