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

Open Access 01-12-2024 | NSCLC | Review

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

Authors: Ting Mei, Ting Wang, Qinghua Zhou

Published in: Clinical and Experimental Medicine | Issue 1/2024

Login to get access

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.
Literature
1.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
7.
go back to reference 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
11.
go back to reference 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.
14.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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
20.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
53.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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
79.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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.
go back to reference 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
Metadata
Title
Multi-omics and artificial intelligence predict clinical outcomes of immunotherapy in non-small cell lung cancer patients
Authors
Ting Mei
Ting Wang
Qinghua Zhou
Publication date
01-12-2024
Publisher
Springer International Publishing
Published in
Clinical and Experimental Medicine / Issue 1/2024
Print ISSN: 1591-8890
Electronic ISSN: 1591-9528
DOI
https://doi.org/10.1007/s10238-024-01324-0

Other articles of this Issue 1/2024

Clinical and Experimental Medicine 1/2024 Go to the issue
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.