Plasma-based microsatellite instability detection strategy to guide immune checkpoint blockade treatment

Zhenghang Wang, Xiaochen Zhao, Chan Gao, Jifang Gong, Xicheng Wang, Jing Gao, Zhongwu Li, Jie Wang, Bo Yang, Lei Wang, Bei Zhang, Yifan Zhou, Dalei Wang, Xiaofang Li, Yuezong Bai, Jian Li, Lin Shen, Zhenghang Wang, Xiaochen Zhao, Chan Gao, Jifang Gong, Xicheng Wang, Jing Gao, Zhongwu Li, Jie Wang, Bo Yang, Lei Wang, Bei Zhang, Yifan Zhou, Dalei Wang, Xiaofang Li, Yuezong Bai, Jian Li, Lin Shen

Abstract

Background: Microsatellite instability (MSI) represents the first pan-cancer biomarker approved to guide immune checkpoint blockade (ICB) treatment. However its widespread testing, especially outside of gastrointestinal cancer, is hampered by tissue availability.

Methods: An algorithm for detecting MSI from peripheral blood was established and validated using clinical plasma samples. Its value for predicting ICB efficacy was evaluated among 60 patients with advanced gastrointestinal cancer. The landscape of MSI in blood was also explored among 5138 advanced solid tumors.

Results: The algorithm included 100 microsatellite markers with high capture efficiency, sensitivity, and specificity. In comparison with orthogonal tissue PCR results, the method displayed a sensitivity of 82.5% (33/40) and a specificity of 96.2% (201/209), for an overall accuracy of 94.0% (234/249). When the clinical validation cohort was dichotomized by pretreatment blood MSI (bMSI), bMSI-high (bMSI-H) predicted both improved progression-free survival and overall survival than the blood microsatellite stable (bMSS) patients (HRs: 0.431 and 0.489, p=0.005 and 0.034, respectively). Four patients with bMSS were identified to have high blood tumor mutational burden (bTMB-H) and trended towards a better survival than the bMSS-bTMB-low (bTMB-L) subset (HR 0.026, 95% CI 0 to 2.635, p=0.011). These four patients with bMSS-bTMB-H plus the bMSI-H group collectively displayed significantly improved survival over the bMSS-bTMB-L patients (HR 0.317, 95% CI 0.157 to 0.640, p<0.001). Pan-cancer prevalence of bMSI-H was largely consistent with that shown for tissue except for much lower rates in endometrial and gastrointestinal cancers, and a remarkably higher prevalence in prostate cancer relative to other cancer types.

Conclusions: We have developed a reliable and robust next generation sequencing-based bMSI detection strategy which, in combination with a panel enabling concurrent profiling of bTMB from a single blood draw, may better inform ICB treatment.

Keywords: biomarkers; gastrointestinal neoplasms; genome instability; immunotherapy; tumor.

Conflict of interest statement

Competing interests: None declared.

© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
A diagram illustrating the study design and samples used for each step. bMSI-H, blood-based MSI high; MS, microsatellite; MSI-H, microsatellite instability high; MSS, microsatellite stable; PD-1, programmed cell death protein-1; PD-L1, programmed death ligand 1.
Figure 2
Figure 2
Cut-off value determination to define the blood microsatellite instability-high (bMSI-H) status. For each sample, a bMSI score was calculated as the fraction of unstable loci in the 100 selected loci. To aim for a specificity of at least 95% according to pairwise comparison with tissue PCR results, the threshold for defining MSI-H was determined to be 0.2 as shown by the dashed line. Red dots represent tissue PCR-defined MSI-H tumors, and blue dots represent tissue PCR-defined (microsatellite stable) MSS tumors.
Figure 3
Figure 3
Best response of target lesions from the initiation of immune checkpoint blockade (ICB) treatment in the bMSI-H (A) and bMSS (B) subgroups. The upper and lower dashed lines mark progressive disease and partial response according to RECIST V.1.1, respectively. bMSI-H, blood-based microsatellite instability high; bMSS, blood microsatellite stable;RECIST, ResponseEvaluation Criteria in Solid Tumors.
Figure 4
Figure 4
Kaplan-Meier analyzes of progression-free survival (PFS) (A) and overall survival (OS) (B) in the patients with bMSI-H versus bMSS. A cohort of 60 patients with advanced gastrointestinal cancer were stratified based on their pretreatment bMSI statuses and the Kaplan-Meier curves for PFS and OS were compared between the bMSI-H and the bMSS subgroups. The tick marks indicate censored data. bMSI-H, blood-based microsatelliteinstability high; bMSS, blood microsatellite stable.
Figure 5
Figure 5
Blood tumor mutational burden (bTMB) as a complement to bMSI to inform ICB treatment. (A) The bTMB levels in the bMSI-H versus the bMSS groups; (B) Kaplan-Meier analysis of OS in the patients with bMSS-TMB-H versus bMSS-TMB-L; (C) Kaplan-Meier analysis of OS in the bMSI-H or bMSI-H versus the bMSS-bTMB-L patients. A cohort of 60 patients with advanced gastrointestinal cancer were stratified based on pretreatment bMSI and bTMB statuses, and those classified as bMSI-H-bTMB-L, bMSS-bTMB-H, or bMSI-H-bTMB-H were pooled as one group in (C). The tick marks indicate censored data. bMSI-H, blood-based microsatellite instability high; bMSS, blood microsatellite stable;H, high; ICB, immune checkpoint blockade; L, low; OS, overallsurvival.
Figure 6
Figure 6
Prevalence of bMSI across 18 cancer types comprizing 5138 solid tumors. bMSI, blood-based microsatellite instability; GIST, gastrointestinal stromal tumor; NSCLC, non-small cell lung cancer.

References

    1. Le DT, Le DT, Diaz LA, Jr. Pd-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med 2015;373:1979. 10.1056/NEJMc1510353
    1. Le DT, Durham JN, Smith KN, et al. . Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017;357:409–13. 10.1126/science.aan6733
    1. Boland CR, Thibodeau SN, Hamilton SR, et al. . A national cancer Institute workshop on microsatellite instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer. Cancer Res 1998;58:5248–57.
    1. Warthin AS. Classics in oncology. heredity with reference to carcinoma as shown by the study of the cases examined in the pathological laboratory of the University of Michigan, 1895-1913. by Aldred Scott Warthin. 1913. CA Cancer J Clin 1985;35:348–59. 10.3322/canjclin.35.6.348
    1. Lynch HT, Shaw MW, Magnuson CW, et al. . Hereditary factors in cancer. study of two large midwestern kindreds. Arch Intern Med 1966;117:206–12.
    1. Benson AB, Arnoletti JP, Bekaii-Saab T, et al. . Colon cancer. J Natl Compr Canc Netw 2011;9:1238–90. 10.6004/jnccn.2011.0104
    1. Benson AB, Venook AP, Al-Hawary MM, et al. . Small bowel adenocarcinoma, version 1.2020, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw 2019;17:1109–33. 10.6004/jnccn.2019.0043
    1. Koh W-J, Abu-Rustum NR, Bean S, et al. . Cervical cancer, version 3.2019, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw 2019;17:64–84. 10.6004/jnccn.2019.0001
    1. Network NCC Gastric cancer, version 5.2017, NCCN clinical practice guidelines in oncology. Available:
    1. Umar A, Boland CR, Terdiman JP, et al. . Revised Bethesda guidelines for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability. J Natl Cancer Inst 2004;96:261–8. 10.1093/jnci/djh034
    1. Goel A, Nagasaka T, Hamelin R, et al. . An optimized pentaplex PCR for detecting DNA mismatch repair-deficient colorectal cancers. PLoS One 2010;5:e9393. 10.1371/journal.pone.0009393
    1. Chen M-L, Chen J-Y, Hu J, et al. . Comparison of microsatellite status detection methods in colorectal carcinoma. Int J Clin Exp Pathol 2018;11:1431–8.
    1. Dudley JC, Lin M-T, Le DT, et al. . Microsatellite instability as a biomarker for PD-1 blockade. Clin Cancer Res 2016;22:813–20. 10.1158/1078-0432.CCR-15-1678
    1. Evrard C, Tachon G, Randrian V, et al. . Microsatellite instability: diagnosis, heterogeneity, discordance, and clinical impact in colorectal cancer. Cancers 2019;11. 10.3390/cancers11101567. [Epub ahead of print: 15 10 2019].
    1. Cohen R, Hain E, Buhard O, et al. . Association of primary resistance to immune checkpoint inhibitors in metastatic colorectal cancer with misdiagnosis of microsatellite instability or mismatch repair deficiency status. JAMA Oncol 2019;5:551–5. 10.1001/jamaoncol.2018.4942
    1. Lu Y, Soong TD, Elemento O. A novel approach for characterizing microsatellite instability in cancer cells. PLoS One 2013;8:e63056. 10.1371/journal.pone.0063056
    1. Kautto EA, Bonneville R, Miya J, et al. . Performance evaluation for rapid detection of pan-cancer microsatellite instability with MANTIS. Oncotarget 2017;8:7452–63. 10.18632/oncotarget.13918
    1. Niu B, Ye K, Zhang Q, et al. . MSIsensor: microsatellite instability detection using paired tumor-normal sequence data. Bioinformatics 2014;30:1015–6. 10.1093/bioinformatics/btt755
    1. Salipante SJ, Scroggins SM, Hampel HL, et al. . Microsatellite instability detection by next generation sequencing. Clin Chem 2014;60:1192–9. 10.1373/clinchem.2014.223677
    1. Zhu L, Huang Y, Fang X, et al. . A novel and reliable method to detect microsatellite instability in colorectal cancer by next-generation sequencing. J Mol Diagn 2018;20:225–31. 10.1016/j.jmoldx.2017.11.007
    1. Hause RJ, Pritchard CC, Shendure J, et al. . Classification and characterization of microsatellite instability across 18 cancer types. Nat Med 2016;22:1342–50. 10.1038/nm.4191
    1. Willis J, Lefterova MI, Artyomenko A, et al. . Validation of microsatellite instability detection using a comprehensive plasma-based genotyping panel. Clin Cancer Res 2019;25:7035–45. 10.1158/1078-0432.CCR-19-1324
    1. Georgiadis A, Durham JN, Keefer LA, et al. . Noninvasive detection of microsatellite instability and high tumor mutation burden in cancer patients treated with PD-1 blockade. Clin Cancer Res 2019;25:7024–34. 10.1158/1078-0432.CCR-19-1372
    1. Cai Z, Wang Z, Liu C, et al. . Detection of Microsatellite Instability from Circulating Tumor DNA by Targeted Deep Sequencing. J Mol Diagn 2020;22:860–70. 10.1016/j.jmoldx.2020.04.210
    1. Du C, Zhao J, Xue W, et al. . Prognostic value of microsatellite instability in sporadic locally advanced rectal cancer following neoadjuvant radiotherapy. Histopathology 2013;62:723–30. 10.1111/his.12069
    1. Gaffney EV, Tsai SC, Dell'Aquila ML, et al. . Production of growth-inhibitory activity in serum-free medium by human monocytic leukemia cells. Cancer Res 1983;43:3668–73.
    1. Wang Z, Duan J, Cai S, et al. . Assessment of blood tumor mutational burden as a potential biomarker for immunotherapy in patients with non-small cell lung cancer with use of a next-generation sequencing cancer gene panel. JAMA Oncol 2019;5:696–702. 10.1001/jamaoncol.2018.7098
    1. Yang N, Li Y, Liu Z, et al. . The characteristics of ctDNA reveal the high complexity in matching the corresponding tumor tissues. BMC Cancer 2018;18:319. 10.1186/s12885-018-4199-7
    1. Mezquita L, Auclin E, Ferrara R, et al. . Association of the lung immune prognostic index with immune checkpoint inhibitor outcomes in patients with advanced non-small cell lung cancer. JAMA Oncol 2018;4:351–7. 10.1001/jamaoncol.2017.4771
    1. Buder-Bakhaya K, Hassel JC. Biomarkers for clinical benefit of immune checkpoint inhibitor Treatment-A review from the melanoma perspective and beyond. Front Immunol 2018;9:1474. 10.3389/fimmu.2018.01474
    1. Petrelli F, Cabiddu M, Coinu A, et al. . Prognostic role of lactate dehydrogenase in solid tumors: a systematic review and meta-analysis of 76 studies. Acta Oncol 2015;54:961–70. 10.3109/0284186X.2015.1043026
    1. Goodman AM, Sokol ES, Frampton GM, et al. . Microsatellite-Stable tumors with high mutational burden benefit from immunotherapy. Cancer Immunol Res 2019;7:1570–3. 10.1158/2326-6066.CIR-19-0149
    1. Timmerman S, Van Rompuy AS, Van Gorp T, et al. . Analysis of 108 patients with endometrial carcinoma using the promise classification and additional genetic analyses for MMR-D. Gynecol Oncol 2020;157:245–51. 10.1016/j.ygyno.2020.01.019
    1. Cancer genome atlas research N. comprehensive molecular characterization of gastric adenocarcinoma. Nature 2014;513:202–9.
    1. Ganesh K, Stadler ZK, Cercek A, et al. . Immunotherapy in colorectal cancer: rationale, challenges and potential. Nat Rev Gastroenterol Hepatol 2019;16:361–75. 10.1038/s41575-019-0126-x
    1. Wang L, Ajani JA. Ushering in liquid biopsy for the microsatellite status: advantages and caveats. Clin Cancer Res 2019;25:6887–9. 10.1158/1078-0432.CCR-19-2585
    1. Kim ST, Cristescu R, Bass AJ, et al. . Comprehensive molecular characterization of clinical responses to PD-1 inhibition in metastatic gastric cancer. Nat Med 2018;24:1449–58. 10.1038/s41591-018-0101-z
    1. Goyal L, Saha SK, Liu LY, et al. . Polyclonal Secondary FGFR2 Mutations Drive Acquired Resistance to FGFR Inhibition in Patients with FGFR2 Fusion-Positive Cholangiocarcinoma. Cancer Discov 2017;7:252–63. 10.1158/-16-1000
    1. Pectasides E, Stachler MD, Derks S, et al. . Genomic heterogeneity as a barrier to precision medicine in gastroesophageal adenocarcinoma. Cancer Discov 2018;8:37–48. 10.1158/-17-0395
    1. Tachon G, Frouin E, Karayan-Tapon L, et al. . Heterogeneity of mismatch repair defect in colorectal cancer and its implications in clinical practice. Eur J Cancer 2018;95:112–6. 10.1016/j.ejca.2018.01.087
    1. He W-Z, Hu W-M, Wang F, et al. . Comparison of mismatch repair status between primary and matched metastatic sites in patients with colorectal cancer. J Natl Compr Canc Netw 2019;17:1174–83. 10.6004/jnccn.2019.7308
    1. Thompson JC, Yee SS, Troxel AB, et al. . Detection of therapeutically targetable driver and resistance mutations in lung cancer patients by next-generation sequencing of cell-free circulating tumor DNA. Clin Cancer Res 2016;22:5772–82. 10.1158/1078-0432.CCR-16-1231
    1. Marcus L, Lemery SJ, Keegan P, et al. . FDA approval summary: pembrolizumab for the treatment of microsatellite Instability-High solid tumors. Clin Cancer Res 2019;25:3753–8. 10.1158/1078-0432.CCR-18-4070
    1. Marabelle A, Le DT, Ascierto PA, et al. . Efficacy of pembrolizumab in patients with Noncolorectal high microsatellite Instability/Mismatch repair-deficient cancer: results from the phase II KEYNOTE-158 study. J Clin Oncol 2020;38:1–10. 10.1200/JCO.19.02105
    1. Le DT, Kim TW, Van Cutsem E, et al. . Phase II open-label study of pembrolizumab in treatment-refractory, microsatellite Instability-High/Mismatch repair-deficient metastatic colorectal cancer: KEYNOTE-164. J Clin Oncol 2020;38:11–19. 10.1200/JCO.19.02107
    1. Abida W, Cheng ML, Armenia J, et al. . Analysis of the prevalence of microsatellite instability in prostate cancer and response to immune checkpoint blockade. JAMA Oncol 2019;5:471–8. 10.1001/jamaoncol.2018.5801
    1. Fraune C, Simon R, Höflmayer D, et al. . High homogeneity of mismatch repair deficiency in advanced prostate cancer. Virchows Arch 2020;476:745–52. 10.1007/s00428-019-02701-x
    1. Ribic CM, Sargent DJ, Moore MJ, et al. . Tumor microsatellite-instability status as a predictor of benefit from fluorouracil-based adjuvant chemotherapy for colon cancer. N Engl J Med 2003;349:247–57. 10.1056/NEJMoa022289

Source: PubMed

3
S'abonner