Cancer Res Treat.  2022 Apr;54(2):424-433. 10.4143/crt.2021.583.

Comparison of the Predictive Power of a Combination versus Individual Biomarker Testing in Non–Small Cell Lung Cancer Patients Treated with Immune Checkpoint Inhibitors

Affiliations
  • 1Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
  • 2Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
  • 3Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea

Abstract

Purpose
Since tumor mutational burden (TMB) and gene expression profiling (GEP) have complementary effects, they may have improved predictive power when used in combination. Here, we investigated the ability of TMB and GEP to predict the immunotherapy response in patients with non–small cell lung cancer (NSCLC) and assessed if this combination can improve predictive power compared to that when used individually.
Materials and Methods
This retrospective cohort study included 30 patients with NSCLC who received immune checkpoint inhibitors (ICI) therapy at the Seoul National University Bundang Hospital. programmed cell death-ligand-1 (PD-L1) protein expression was assessed using immunohistochemistry, and TMB was measured by targeted deep sequencing. Gene expression was determined using NanoString nCounter analysis for the PanCancer IO360 panel, and enrichment analysis were performed.
Results
Eleven patients (36.7%) showed a durable clinical benefit (DCB), whereas 19 (63.3%) showed no durable benefit (NDB). TMB and enrichment scores (ES) showed significant differences between the DCB and NDB groups (p=0.044 and p=0.017, respectively); however, no significant correlations were observed among TMB, ES, and PD-L1. ES was the best single biomarker for predicting DCB (area under the curve [AUC], 0.794), followed by TMB (AUC, 0.679) and PD-L1 (AUC, 0.622). TMB and ES showed the highest AUC (0.837) among other combinations (AUC [TMB and PD-L1], 0.777; AUC [PD-L1 and ES], 0.763) and was similar to that of all biomarkers used together (0.832).
Conclusion
The combination of TMB and ES may be an effective predictive tool to identify patients with NSCLC patients who would possibly benefit from ICI therapies.

Keyword

Tumor mutational burden; Gene expression profiling; Predictive biomarker; Immune checkpoint inhibitors; Non/small cell lung cancer

Figure

  • Fig. 1 Summary of the clinical and molecular features associated with response to anti–PD-1/PD-L1 therapy in non-small cell lung cancer patients. Individual patients are represented in each column and sorted according to treatment response (DCB vs. NDB). Tumor histology and smoking status are characterized. PD-L1 expression is stratified as < 1%, 1%–49%, and ≥ 50%. The frequency of a selected gene mutation and tumor mutational burden (mutations/megabase) are sequentially displayed on the histogram. DCB, durable clinical benefit; NDB, no durable benefit; PD-1, programmed death-1; PD-L1, programmed cell death-ligand-1; TMB, tumor mutational burden; VUS, variant of uncertain significance.

  • Fig. 2 Tumor mutational burden (TMB) according to response to anti–PD-1/PD-L1 therapy. TMB was greater in patients with DCB than in those with NDB and was significantly different among those with CR/PR vs. SD vs. progressive disease. Box plots represent medians and interquartile ranges. Vertical lines extend to the 95th percentiles. CR, complete response; DCB, durable clinical benefit; NDB, no durable benefit; PD, progressive disease; PD-1, programmed death-1; PD-L1, programmed cell death-ligand-1; PR, partial response; SD, stable disease; TMB, tumor mutational burden. *p < 0.05.

  • Fig. 3 (A) Top ten pathways with gene ontology molecular function, as identified by singular enrichment analysis. (B) Representative differentially expressed genes in the DCB and NDB groups. (C) Gene set enrichment analysis showed that the DCB group was significantly enriched in the GSE37605 (Treg and Tconv Cells) and GSE20366 (TregLP vs. TconvLP up). DCB, durable clinical benefit; NDB, no durable benefit.

  • Fig. 4 Receiver operating characteristic curve of sensitivity vs. 1-specificity of durable clinical benefit for TMB, ES, PD-L1, and the combination of two or more biomarkers. ES, enrichment score; PD-L1, programmed cell death-ligand-1; TMB, tumor mutational burden.

  • Fig. 5 Kaplan-Meier survival curves showing progression-free survival according to TMB (A) and ES (B). ES, enrichment score; TMB, tumor mutational burden.


Reference

References

1. Brahmer J, Reckamp KL, Baas P, Crino L, Eberhardt WE, Poddubskaya E, et al. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med. 2015; 373:123–35.
Article
2. Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 2015; 373:1627–39.
Article
3. Herbst RS, Baas P, Kim DW, Felip E, Perez-Gracia JL, Han JY, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet. 2016; 387:1540–50.
Article
4. Rittmeyer A, Barlesi F, Waterkamp D, Park K, Ciardiello F, von Pawel J, 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. Lancet. 2017; 389:255–65.
Article
5. Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, et al. Cancer immunology: mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015; 348:124–8.
Article
6. Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med. 2017; 377:2500–1.
Article
7. Hellmann MD, Callahan MK, Awad MM, Calvo E, Ascierto PA, Atmaca A, et al. Tumor mutational burden and efficacy of nivolumab monotherapy and in combination with ipilimumab in small-cell lung cancer. Cancer Cell. 2018; 33:853–61.
Article
8. Goodman AM, Kato S, Bazhenova L, Patel SP, Frampton GM, Miller V, et al. Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol Cancer Ther. 2017; 16:2598–608.
Article
9. Hellmann MD, Paz-Ares L, Bernabe Caro R, Zurawski B, Kim SW, Carcereny Costa E, et al. Nivolumab plus ipilimumab in advanced non-small-cell lung cancer. N Engl J Med. 2019; 381:2020–31.
Article
10. Auslander N, Zhang G, Lee JS, Frederick DT, Miao B, Moll T, et al. Robust prediction of response to immune checkpoint blockade therapy in metastatic melanoma. Nat Med. 2018; 24:1545–9.
Article
11. Jiang P, Gu S, Pan D, Fu J, Sahu A, Hu X, et al. Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nat Med. 2018; 24:1550–8.
Article
12. Yu Y, Zeng D, Ou Q, Liu S, Li A, Chen Y, et al. Association of survival and immune-related biomarkers with immunotherapy in patients with non-small cell lung xancer: a meta-analysis and individual patient-level analysis. JAMA Netw Open. 2019; 2:e196879.
13. Lee JS, Ruppin E. Multiomics prediction of response rates to therapies to inhibit programmed cell death 1 and programmed cell death 1 ligand 1. JAMA Oncol. 2019; 5:1614–8.
Article
14. Lu S, Stein JE, Rimm DL, Wang DW, Bell JM, Johnson DB, et al. Comparison of biomarker modalities for predicting response to PD-1/PD-L1 checkpoint blockade: a systematic review and meta-analysis. JAMA Oncol. 2019; 5:1195–204.
Article
15. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009; 45:228–47.
Article
16. Hernandez-Prera JC, Valderrabano P, Creed JH, de la Iglesia JV, Slebos RJC, Centeno BA, et al. Molecular determinants of thyroid nodules with indeterminate cytology and RAS mutations. Thyroid. 2021; 31:36–49.
Article
17. Reich M, Liefeld T, Gould J, Lerner J, Tamayo P, Mesirov JP. GenePattern 2.0. Nat Genet. 2006; 38:500–1.
Article
18. Kim H, Kwon HJ, Han YB, Park SY, Kim ES, Kim SH, et al. Increased CD3+ T cells with a low FOXP3+/CD8+ T cell ratio can predict anti-PD-1 therapeutic response in non-small cell lung cancer patients. Mod Pathol. 2019; 32:367–75.
Article
19. Ott PA, Bang YJ, Piha-Paul SA, Razak ARA, Bennouna J, Soria JC, 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; 37:318–27.
Article
20. Feng YY, Griffith OL, Griffith M. Clinical implications of neoepitope landscapes for adult and pediatric cancers. Genome Med. 2017; 9:77.
Article
21. Turajlic S, Litchfield K, Xu H, Rosenthal R, McGranahan N, Reading JL, et al. Insertion-and-deletion-derived tumour-specific neoantigens and the immunogenic phenotype: a pan-cancer analysis. Lancet Oncol. 2017; 18:1009–21.
Article
22. McGrail DJ, Pilie PG, Rashid NU, Voorwerk L, Slagter M, Kok M, et al. High tumor mutation burden fails to predict immune checkpoint blockade response across all cancer types. Ann Oncol. 2021; 32:661–72.
Article
23. Dees S, Ganesan R, Singh S, Grewal IS. Regulatory T cell targeting in cancer: emerging strategies in immunotherapy. Eur J Immunol. 2021; 51:280–91.
Article
24. Paluskievicz CM, Cao X, Abdi R, Zheng P, Liu Y, Bromberg JS. T regulatory cells and priming the suppressive tumor microenvironment. Front Immunol. 2019; 10:2453.
Article
25. Yarchoan M, Albacker LA, Hopkins AC, Montesion M, Murugesan K, Vithayathil TT, et al. PD-L1 expression and tumor mutational burden are independent biomarkers in most cancers. JCI Insight. 2019; 4:e126908.
Article
26. Gainor JF, Shaw AT, Sequist LV, Fu X, Azzoli CG, Piotrowska Z, et al. EGFR mutations and ALK rearrangements are associated with low response rtes to PD-1 pathway blockade in non-small cell lung cancer: a retrospective analysis. Clin Cancer Res. 2016; 22:4585–93.
27. Dong ZY, Zhong WZ, Zhang XC, Su J, Xie Z, Liu SY, 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; 23:3012–24.
28. Skoulidis F, Goldberg ME, Greenawalt DM, Hellmann MD, Awad MM, Gainor JF, et al. STK11/LKB1 mutations and PD-1 inhibitor resistance in KRAS-mutant lung adenocarcinoma. Cancer Discov. 2018; 8:822–35.
29. Papillon-Cavanagh S, Doshi P, Dobrin R, Szustakowski J, Walsh AM. STK11 and KEAP1 mutations as prognostic biomarkers in an observational real-world lung adenocarcinoma cohort. ESMO Open. 2020; 5:e000706.
Article
Full Text Links
  • CRT
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr