Cancer Res Treat.  2021 Oct;53(4):973-982. 10.4143/crt.2020.798.

Pan-cancer Analysis of Tumor Mutational Burden and Homologous Recombination DNA Damage Repair Using Targeted Next-Generation Sequencing

Affiliations
  • 1Department of Pathology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
  • 2Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
  • 3Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
  • 4Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, Guangzhou, China
  • 5Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, China
  • 6Department of Ultrasound and Electrocardiogram, Sun Yat-Sen University Cancer Center, Guangzhou, China

Abstract

Purpose
Current variability in methods for tumor mutational burden (TMB) estimation and reporting demonstrates the urgent need for a homogeneous TMB assessment approach. Here, we compared TMB distributions in different cancer types using two customized targeted panels commonly used in clinical practice.
Materials and Methods
TMB spectra of 295- and 1021-gene panels in multiple cancer types were compared using targeted next-generation sequencing (NGS). The TMB distributions across a diverse cohort of 2,332 cancer cases were then investigated for their associations with clinical features. Treatment response data were collected for 222 patients who received immune-checkpoint inhibitors (ICIs) and their homologous recombination DNA damage repair (HR-DDR) and programmed death-ligand 1 (PD-L1) expression were additionally assessed and compared with the TMB and response rate.
Results
The median TMB between gene panels was similar despite a wide range in TMB values. The highest TMB was 8 and 10 in patients with squamous cell carcinoma and esophageal carcinoma according to the classification of histopathology and cancer types, respectively. Twenty-three out of 103 patients (22.3%) were HR-DDR‒positive and could benefit from ICI therapy; out of those 23 patients, seven patients had high TMB (p=0.004). Additionally, PD-L1 expression was not associated with TMB or treatment response among patients receiving ICIs.
Conclusion
Targeted NGS assays demonstrated the ability to evaluate TMB in pan-cancer samples as a tool to predict response to ICIs. In addition, TMB integrated with HR-DDR‒positive status could be a significant biomarker for predicting ICI response in patients.

Keyword

Tumor mutational burden; Homologous recombination DNA damage repair; Pan-cancer; Immunotherapy

Figure

  • Fig. 1 Flowchart illustrating the analytic workflow of the study comprising of TMB calculations from the 295- and 1021-customized sequencing panels. HR-DDR, homologous recombination DNA damage repair; NGS, next-generation sequencing; TMB, tumor mutational burden.

  • Fig. 2 The number of patients was calculated using the 295- and 1021-gene panels, and defined by cancer types (A) and pathological subtypes (B), respectively. SCC, squamous cell carcinoma. Unknown cancer types denote the sites of primary tumor were not available.

  • Fig. 3 TMB distributions across the different cancer types and pathological subtypes. Overview of the median TMB (log10-transformed) across different cancers (A) and pathological types (C). The comparison of TMB value (log10-transformed) defined by two different gene panels in cancers (B) and pathological types (D). SCC, squamous cell carcinoma; TMB, tumor mutational burden.

  • Fig. 4 The efficacy associated with TMB, HR-DDR mutation status PD-L1 expression among patients who received ICI treatment. DCB, durable clinical benefit; HR-DDR, homologous recombination DNA damage repair; ICIs, immunotherapy inhibitors; NDB, no durable benefit; PD-L1, programmed death ligand-1; TMB, tumor mutational burden.


Reference

References

1. Jiang T, Shi T, Zhang H, Hu J, Song Y, Wei J, et al. Tumor neoantigens: from basic research to clinical applications. J Hematol Oncol. 2019; 12:93.
Article
2. Chen L, Ashe S, Brady WA, Hellstrom I, Hellstrom KE, Ledbetter JA, et al. Costimulation of antitumor immunity by the B7 counterreceptor for the T lymphocyte molecules CD28 and CTLA-4. Cell. 1992; 71:1093–102.
Article
3. Leach DR, Krummel MF, Allison JP. Enhancement of antitumor immunity by CTLA-4 blockade. Science. 1996; 271:1734–6.
Article
4. Larsen TV, Hussmann D, Nielsen AL. PD-L1 and PD-L2 expression correlated genes in non-small-cell lung cancer. Cancer Commun (Lond). 2019; 39:30.
Article
5. Hirano F, Kaneko K, Tamura H, Dong H, Wang S, Ichikawa M, et al. Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity. Cancer Res. 2005; 65:1089–96.
6. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012; 366:2443–54.
Article
7. Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, et al. Nivolumab versus everolimus in advanced renal-cell carcinoma. N Engl J Med. 2015; 373:1803–13.
Article
8. Bracarda S, Altavilla A, Hamzaj A, Sisani M, Marrocolo F, Del Buono S, et al. Immunologic checkpoints blockade in renal cell, prostate, and urothelial malignancies. Semin Oncol. 2015; 42:495–505.
Article
9. Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012; 366:2455–65.
Article
10. Reck M, Rodriguez-Abreu D, Robinson AG, Hui R, Csoszi T, Fulop A, et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N Engl J Med. 2016; 375:1823–33.
Article
11. Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017; 357:409–13.
12. Folprecht G. Tumor mutational burden as a new biomarker for PD-1 antibody treatment in gastric cancer. Cancer Commun (Lond). 2019; 39:74.
Article
13. Shekarian T, Valsesia-Wittmann S, Brody J, Michallet MC, Depil S, Caux C, et al. Pattern recognition receptors: immune targets to enhance cancer immunotherapy. Ann Oncol. 2017; 28:1756–66.
Article
14. Li A, Yang JJ, Zhang XC, Zhang Z, Su J, Gou LY, et al. Acquired MET Y1248H and D1246N mutations mediate resistance to MET inhibitors in non-small cell lung cancer. Clin Cancer Res. 2017; 23:4929–37.
Article
15. Buchhalter I, Rempel E, Endris V, Allgauer M, Neumann O, Volckmar AL, et al. Size matters: Dissecting key parameters for panel-based tumor mutational burden analysis. Int J Cancer. 2019; 144:848–58.
Article
16. Heeke AL, Pishvaian MJ, Lynce F, Xiu J, Brody JR, Chen WJ, et al. Prevalence of homologous recombination-related gene mutations across multiple cancer types. JCO Precis Oncol. 2018; 2018:PO.1700286.
Article
17. Rimm DL, Han G, Taube JM, Yi ES, Bridge JA, Flieder DB, et al. A prospective, multi-institutional, pathologist-based assessment of 4 immunohistochemistry assays for PD-L1 expression in non-small cell lung cancer. JAMA Oncol. 2017; 3:1051–8.
Article
18. Campesato LF, Barroso-Sousa R, Jimenez L, Correa BR, Sabbaga J, Hoff PM, et al. Comprehensive cancer-gene panels can be used to estimate mutational load and predict clinical benefit to PD-1 blockade in clinical practice. Oncotarget. 2015; 6:34221–7.
Article
19. Ma F, Guan Y, Yi Z, Chang L, Li Q, Chen S, et al. Assessing tumor heterogeneity using ctDNA to predict and monitor therapeutic response in metastatic breast cancer. Int J Cancer. 2020; 146:1359–68.
Article
20. Wang Y, Zhao C, Chang L, Jia R, Liu R, Zhang Y, et al. Circulating tumor DNA analyses predict progressive disease and indicate trastuzumab-resistant mechanism in advanced gastric cancer. EBioMedicine. 2019; 43:261–9.
Article
21. Zhou Z, Zhu L, Niu X, Shen S, Zhao Y, Zhang J, et al. Comparison of genomic landscapes of large cell neuroendocrine carcinoma, small cell lung carcinoma, and large cell carcinoma. Thorac Cancer. 2019; 10:839–47.
Article
22. Zhang Y, Chang L, Yang Y, Fang W, Guan Y, Wu A, et al. The correlations of tumor mutational burden among single-region tissue, multi-region tissues and blood in non-small cell lung cancer. J Immunother Cancer. 2019; 7:98.
Article
23. Sun S, Liu Y, Eisfeld AK, Zhen F, Jin S, Gao W, et al. Identification of germline mismatch repair gene mutations in lung cancer patients with paired tumor-normal next generation sequencing: a retrospective study. Front Oncol. 2019; 9:550.
Article
24. Singal G, Miller PG, Agarwala V, Li G, Kaushik G, Backenroth D, 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; 321:1391–9.
Article
25. Yao Y, Zhang T, Qi L, Liu R, Liu G, Wang X, et al. Competitive endogenous RNA network construction and comparison of lung squamous cell carcinoma in smokers and nonsmokers. Dis Markers. 2019; 2019:5292787.
Article
26. Wang K, McDermott JD, Schrock AB, Elvin JA, Gay L, Karam SD, et al. Comprehensive genomic profiling of salivary mucoepidermoid carcinomas reveals frequent BAP1, PIK3CA, and other actionable genomic alterations. Ann Oncol. 2017; 28:748–53.
27. Chalmers ZR, Connelly CF, Fabrizio D, Gay L, Ali SM, Ennis R, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017; 9:34.
Article
28. Salem ME, Puccini A, Grothey A, Xiu J, Goldberg R, Kim ES, et al. Comparative molecular analysis between microsatellite instability-high (MSI-H) tumors with high tumor mutational burden (TMB-H) versus MSI-H tumors with TMB-intermediate/low. Ann Oncol. 2018; 29(Suppl 8):VIII649–69.
Article
29. Pursell ZF, Isoz I, Lundstrom EB, Johansson E, Kunkel TA. Yeast DNA polymerase epsilon participates in leading-strand DNA replication. Science. 2007; 317:127–30.
30. Petitjean A, Mathe E, Kato S, Ishioka C, Tavtigian SV, Hainaut P, et al. Impact of mutant p53 functional properties on TP53 mutation patterns and tumor phenotype: lessons from recent developments in the IARC TP53 database. Hum Mutat. 2007; 28:622–9.
31. Yang S, Yu X, Fan Y, Shi X, Jin Y. Clinicopathologic characteristics and survival outcome in patients with advanced lung adenocarcinoma and KRAS mutation. J Cancer. 2018; 9:2930–7.
Article
32. Telli ML, Timms KM, Reid J, Hennessy B, Mills GB, Jensen KC, et al. Homologous Recombination Deficiency (HRD) score predicts response to platinum-containing neoadjuvant chemotherapy in patients with triple-negative breast cancer. Clin Cancer Res. 2016; 22:3764–73.
Article
33. Teo MY, Seier K, Ostrovnaya I, Regazzi AM, Kania BE, Moran MM, et al. Alterations in DNA damage response and repair genes as potential marker of clinical benefit from PD-1/PD-L1 blockade in advanced urothelial cancers. J Clin Oncol. 2018; 36:1685–94.
Article
34. Kim SJ, Kim S, Kim DW, Kim M, Keam B, Kim TM, et al. Alterations in PD-L1 expression associated with acquisition of resistance to ALK inhibitors in ALK-rearranged lung cancer. Cancer Res Treat. 2019; 51:1231–40.
Article
35. Rosenberg JE, Hoffman-Censits J, Powles T, van der Heijden MS, Balar AV, Necchi A, et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet. 2016; 387:1909–20.
36. Labriola MK, Zhu J, Gupta R, McCall S, Jackson J, Kong EF, et al. Characterization of tumor mutation burden, PD-L1 and DNA repair genes to assess relationship to immune checkpoint inhibitors response in metastatic renal cell carcinoma. J Immunother Cancer. 2020; 8:e000319.
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