The combination of tumor mutational burden and T-cell receptor repertoire predicts the response to immunotherapy in patients with advanced non–small cell lung cancer

Yalun Li1,2, Liyan Ji3, Yingqian Zhang3, Jiexin Zhang4, Alexandre Reuben5, Hao Zeng6, Qin Huang6, Qi Wei6, Sihan Tan6, Xuefeng Xia3, Weimin Li1, Jianjun Zhang5,7,8,9(), Panwen Tian1,2()

PDF
MedComm ›› 2024, Vol. 5 ›› Issue (6) : e604. DOI: 10.1002/mco2.604
ORIGINAL ARTICLE

The combination of tumor mutational burden and T-cell receptor repertoire predicts the response to immunotherapy in patients with advanced non–small cell lung cancer

  • Yalun Li1,2, Liyan Ji3, Yingqian Zhang3, Jiexin Zhang4, Alexandre Reuben5, Hao Zeng6, Qin Huang6, Qi Wei6, Sihan Tan6, Xuefeng Xia3, Weimin Li1, Jianjun Zhang5,7,8,9(), Panwen Tian1,2()
Author information +
History +

Abstract

Tumor mutational burden (TMB) and T-cell receptor (TCR) might predict the response to immunotherapy in patients with non–small cell lung cancer (NSCLC). However, the predictive value of the combination of TMB and TCR was not clear. Targeted DNA and TCR sequencing were performed on tumor biopsy specimens. We combined TMB and TCR diversity into a TMB-and-TCR (TMR) score using logistic regression. In total, 38 patients with advanced NSCLC were divided into a discovery set (n = 17) and validation set (n = 21). A higher TMR score was associated with better response and longer progression-free survival to immunotherapy in both the discovery set and validation set. The performance of TMR score was confirmed in the two external validation cohorts of 225 NSCLC patients and 306 NSCLC patients. Tumors with higher TMR scores were more likely to combine with LRP1B gene mutation (p = 0.027) and top 1% CDR3 sequences (p = 0.001). Furthermore, LRP1B allele frequency was negatively correlated with the top 1% CDR3 sequences (r = –0.55, p = 0.033) and positively correlated with tumor shrinkage (r = 0.68, p = 0.007). The TMR score could serve as a potential predictive biomarker for the response to immunotherapy in advanced NSCLC.

Keywords

advanced non–small cell lung cancer / clonality / immunotherapy / T-cell receptors / tumor mutational burden

Cite this article

Download citation ▾
Yalun Li, Liyan Ji, Yingqian Zhang, Jiexin Zhang, Alexandre Reuben, Hao Zeng, Qin Huang, Qi Wei, Sihan Tan, Xuefeng Xia, Weimin Li, Jianjun Zhang, Panwen Tian. The combination of tumor mutational burden and T-cell receptor repertoire predicts the response to immunotherapy in patients with advanced non–small cell lung cancer. MedComm, 2024, 5(6): e604 https://doi.org/10.1002/mco2.604

References

1 JJ Havel, D Chowell, TA Chan. The evolving landscape of biomarkers for checkpoint inhibitor immunotherapy. Nat Rev Cancer. 2019;19(3):133-150. doi:
2 S Wang, Z He, X Wang, et al. Antigen presentation and tumor immunogenicity in cancer immunotherapy response prediction. eLife. 2019;8:e49020. doi:
3 M Ayers, J Lunceford, M Nebozhyn, et al. IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest. 2017;127(8):2930-2940. doi:
4 RM Samstein, CH Lee, AN Shoushtari, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet. 2019;51(2):202-206. doi:
5 DJ McGrail, PG Pilié, NU Rashid, et al. High tumor mutation burden fails to predict immune checkpoint blockade response across all cancer types. Ann Oncol. 2021;32(5):661-672. doi:
6 MD Hellmann, T Nathanson, H Rizvi, et al. Genomic Features of response to combinaion immunotherapy in patients with advanced non-small-cell lung cancer. Cancer Cell. 2018;33(5):843-852. doi:
7 R Cristescu, R Mogg, M Ayers, et al. Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science. 2018;362(6411):eaar3593. doi:
8 LM Sholl, FR Hirsch, D Hwang, et al. The promises and challenges of tumor mutation burden as an immunotherapy biomarker: a perspective from the International Association for the Study of Lung Cancer Pathology Committee. J Thorac Oncol. 2020;15(9):1409-1424. doi:
9 M Yarchoan, LA Albacker, AC Hopkins, et al. PD-L1 expression and tumor mutational burden are independent biomarkers in most cancers. JCI insight. 2019;4(6):e126908. doi:
10 Y Chen, Y Wang, H Luo, et al. The frequency and inter-relationship of PD-L1 expression and tumour mutational burden across multiple types of advanced solid tumours in China. Exp Hematol Oncol. 2020;9:17. doi:
11 K Li, Z Yuan, J Lyu, et al. PD-1 suppresses TCR-CD8 cooperativity during T-cell antigen recognition. Nat Commun. 2021;12(1):2746. doi:
12 J Zhang, Z Ji, JX Caushi, et al. Compartmental analysis of T-cell clonal dynamics as a function of pathologic response to neoadjuvant PD-1 blockade in resectable non-small cell lung cancer. Clin Cancer Res. 2020;26(6):1327-1337. doi:
13 M Casarrubios, A Cruz-Bermúdez, E Nadal, et al. Pretreatment tissue TCR repertoire evenness is associated with complete pathologic response in patients with NSCLC receiving neoadjuvant chemoimmunotherapy. Clin Cancer Res. 2021;27(21):5878-5890. doi:
14 Q Zhang, Q Jia, T Deng, et al. Heterogeneous expansion of CD4+ tumor-infiltrating T-lymphocytes in clear cell renal cell carcinomas. Biochem Biophys Res Commun. 2015;458(1):70-76. doi:
15 K Nakanishi, Y Kukita, H Segawa, et al. Characterization of the T-cell receptor beta chain repertoire in tumor-infiltrating lymphocytes. Cancer Med. 2016;5(9):2513-2521. doi:
16 T Cukalac, WT Kan, P Dash, et al. Paired TCRalphabeta analysis of virus-specific CD8(+) T cells exposes diversity in a previously defined ‘narrow’ repertoire. Immunol Cell Biol. 2015;93(9):804-814. doi:
17 P Dash, AJ Fiore-Gartland, T Hertz, et al. Quantifiable predictive features define epitope-specific T cell receptor repertoires. Nature. 2017;547(7661):89-93. doi:
18 H Huang, C Wang, F Rubelt, et al. Analyzing the Mycobacterium tuberculosis immune response by T-cell receptor clustering with GLIPH2 and genome-wide antigen screening. Nat Biotechnol. 2020;38(10):1194-1202. doi:
19 J Glanville, H Huang, A Nau, et al. Identifying specificity groups in the T cell receptor repertoire. Nature. 2017;547(7661):94-98. doi:
20 J Wu, AH Pendegraft, M Byrne-Steele, et al. Expanded TCRbeta CDR3 clonotypes distinguish Crohn's disease and ulcerative colitis patients. Mucosal Immunol. 2018;11(5):1487-1495. doi:
21 L Werner, MY Nunberg, E Rechavi, et al. Altered T cell receptor beta repertoire patterns in pediatric ulcerative colitis. Clin Exp Immunol. 2019;196(1):1-11. doi:
22 MJ Peak, JG Peak, CM Blaumueller, et al. Photosensitized DNA breaks and DNA-to-protein crosslinks induced in human cells by antitumor agent gilvocarcin V. Chem Biol Interact. 1988;67(3-4):267-274. doi:
23 DA Yohannes, TL Freitag, A de Kauwe, et al. Deep sequencing of blood and gut T-cell receptor beta-chains reveals gluten-induced immune signatures in celiac disease. Sci Rep. 2017;7(1):17977. doi:
24 HR Seay, E Yusko, SJ Rothweiler, et al. Tissue distribution and clonal diversity of the T and B cell repertoire in type 1 diabetes. JCI insight. 2016;1(20):e88242. doi:
25 LC Brown, MD Tucker, R Sedhom, et al. LRP1B mutations are associated with favorable outcomes to immune checkpoint inhibitors across multiple cancer types. J Immunother Cancer. 2021;9(3):e001792. doi:
26 HA Shihab, MF Rogers, J Gough, et al. An integrative approach to predicting the functional effects of non-coding and coding sequence variation. Bioinformatics. 2015;31(10):1536-1543. doi:
27 D Wang, B Elenbaas, K Murugesan, et al. Relationship among DDR gene mutations, TMB and PD-L1 in solid tumour genomes identified using clinically actionable biomarker assays. NPJ Precis Oncol. 2023;7(1):103. doi:
28 A Reuben, J Zhang, SH Chiou, et al. Comprehensive T cell repertoire characterization of non-small cell lung cancer. Nat Commun. 2020;11(1):603. doi:
29 QJ Wang, Z Yu, K Griffith, et al. Identification of T-cell receptors targeting KRAS-mutated human tumors. Cancer Immunol Res. 2016;4(3):204-214. doi:
30 KR Lin, DM Pang, YB Jin, et al. Circulating CD8(+) T-cell repertoires reveal the biological characteristics of tumors and clinical responses to chemotherapy in breast cancer patients. Cancer Immunol Immunother. 2018;67(11):1743-1752. doi:
31 M Zheng, X Zhang, Y Zhou, et al. TCR repertoire and CDR3 motif analyses depict the role of alphabeta T cells in Ankylosing spondylitis. EBioMedicine. 2019;47:414-426. doi:
32 Cancer Genome Atlas Research N. Comprehensive genomic characterization of squamous cell lung cancers. Nature. 2012;489(7417):519-525. doi:
33 J Chen, H Yang, ASM Teo, et al. Genomic landscape of lung adenocarcinoma in East Asians. Nat Genet. 2020;52(2):177-186. doi:
34 D Nejman, I Livyatan, G Fuks, et al. The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Science. 2020;368(6494):973-980. doi:
35 H Chen, W Chong, Q Wu, et al. Association of LRP1B mutation with tumor mutation burden and outcomes in melanoma and non-small cell lung cancer patients treated with immune check-point blockades. Front Immunol. 2019;10:1113. doi:
36 C Ye, W Chong, Y Liu, et al. Suppression of tumorigenesis in LUAD by LRP1B through regulation of the IL-6-JAK-STAT3 pathway. Am J Cancer Res. 2023;13(7):2886-2905.
37 T Matsuda, E Miyauchi, YW Hsu, et al. TCR sequencing analysis of cancer tissues and tumor draining lymph nodes in colorectal cancer patients. Oncoimmunology. 2019;8(6):e1588085. doi:
38 Consortium ITP-CAoWG. Pan-cancer analysis of whole genomes. Nature. 2020;578(7793):82-93. doi:
39 L Seymour, J Bogaerts, A Perrone, et al. iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol. 2017;18(3):e143-e152. doi:
40 P Tian, H Zeng, L Ji, et al. Lung adenocarcinoma with ERBB2 exon 20 insertions: comutations and immunogenomic features related to chemoimmunotherapy. Lung Cancer. 2021;160:50-58. doi:
41 Q Jia, W Wu, Y Wang, et al. Local mutational diversity drives intratumoral immune heterogeneity in non-small cell lung cancer. Nat Commun. 2018;9(1):5361. doi:
42 A Ravi, MD Hellmann, MB Arniella, et al. Genomic and transcriptomic analysis of checkpoint blockade response in advanced non-small cell lung cancer. Nat Genet. 2023;55(5):807-819. doi:
43 P Jiang, S Gu, D Pan, et al. Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nat Med. 2018;24(10):1550-1558. doi:
PDF

Accesses

Citations

Detail

Sections
Recommended

/