Tumor mutational burden predicts the efficacy of pembrolizumab monotherapy: a pan-tumor retrospective analysis of participants with advanced solid tumors

Razvan Cristescu, Deepti Aurora-Garg, Andrew Albright, Lei Xu, Xiao Qiao Liu, Andrey Loboda, Lixin Lang, Fan Jin, Eric H Rubin, Alexandra Snyder, Jared Lunceford, Razvan Cristescu, Deepti Aurora-Garg, Andrew Albright, Lei Xu, Xiao Qiao Liu, Andrey Loboda, Lixin Lang, Fan Jin, Eric H Rubin, Alexandra Snyder, Jared Lunceford

Abstract

Background: Several studies have evaluated the relationship between tumor mutational burden (TMB) and outcomes of immune checkpoint inhibitors. In the phase II KEYNOTE-158 study of pembrolizumab monotherapy for previously treated recurrent or metastatic cancer, high TMB as assessed by the FoundationOne CDx was associated with an improved objective response rate (ORR).

Methods: We retrospectively assessed the relationship between TMB and efficacy in participants with previously treated advanced solid tumors enrolled in 12 trials that evaluated pembrolizumab monotherapy, including 3 randomized trials that compared pembrolizumab with chemotherapy. TMB was assessed in formalin-fixed, paraffin-embedded pretreatment tumor samples by whole-exome sequencing. High TMB was defined as ≥175 mutations/exome. Microsatellite instability (MSI) phenotype was based on whole-exome sequencing results. Programmed death ligand 1 (PD-L1) expression was assessed by immunohistochemistry. The primary end point was ORR assessed per RECIST V.1.1 by independent central review. Other end points included progression-free survival (PFS) assessed per RECIST V.1.1 by independent central review and overall survival (OS).

Results: Of the 2234 participants in the analysis, 1772 received pembrolizumab monotherapy and 462 received chemotherapy. Among the pembrolizumab-treated participants, ORR was 31.4% (95% CI 27.1 to 36.0) in the 433 participants with TMB ≥175 mutations/exome and 9.5% (95% CI 8.0 to 11.2) in the 1339 participants with TMB <175 mutations/exome. The association of TMB with ORR was observed regardless of PD-L1 expression and not driven by specific tumor types or participants with very high TMB or high MSI. In the 3 randomized controlled trials, TMB was associated with ORR (p≤0.016), PFS (p≤0.005), and OS (p≤0.029) of pembrolizumab but not of chemotherapy (p≥0.340, p≥0.643, and p≥0.174, respectively), and pembrolizumab improved efficacy versus chemotherapy in participants with TMB ≥175 mutations/exome.

Conclusions: TMB ≥175 mutations/exome is associated with clinically meaningful improvement in the efficacy of pembrolizumab monotherapy and improved outcomes for pembrolizumab versus chemotherapy across a wide range of previously treated advanced solid tumor types. These data suggest TMB has broad clinical utility irrespective of tumor type, PD-L1 expression, or MSI status and support its use as a predictive biomarker for pembrolizumab monotherapy in participants with previously treated advanced solid tumors.

Trial registration: ClinicalTrials.gov NCT01295827 NCT01704287 NCT01905657 NCT01848834 NCT02054806 NCT02256436 NCT02255097 NCT02335411 NCT02370498 NCT02447003 NCT02674061 NCT02787005.

Keywords: immunotherapy; programmed cell death 1 receptor; tumor biomarkers.

Conflict of interest statement

Competing interests: RC, DA-G, AA, LX, AL, LL, FJ, EHR, and JL are full-time employees of Merck Sharp & Dohme Corp, a subsidiary of Merck & Co, Inc, Kenilworth, New Jersey, USA and hold stock in Merck & Co, Inc, Kenilworth, New Jersey, USA. XQL is a full-time employee of MSD China. AS was a full-time employee of Merck Sharp & Dohme Corp, a subsidiary of Merck & Co, Inc, Kenilworth, New Jersey, USA at the time the study was conducted.

© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Validation of the optimal cutpoint for TMB assessed by WES. (A) Comparison of inflammation in the tumor microenvironment assessed using an 18-gene T-cell inflamed GEP in participants with TMB above versus below potential WES cutpoints for mutational burden. P values are two-sided and assessed using the Wilcoxon rank-sum test. The vertical dotted line represents TMB 175 mut/exome. (B) The optimal TMB cut-off assessed by WES that corresponds with the FoundationOne CDx TMB cutpoint of 10 mut/megabase. The Youden index of 175 mut/exome assessed per WES is the point of maximal average positive and negative agreement rates with 10 mut/megabase by the FoundationOne CDx. The area under the receiver-operating characteristic curve is 0.92. GEP, gene expression profile; mut, mutations; TMB, tumor mutational burden; WES, whole-exome sequencing.
Figure 2
Figure 2
Proportion of participants in the pooled pembrolizumab population with whole-exome sequencing TMB ≥175 mut/exome and TMB

Figure 3

Objective response rate in participants…

Figure 3

Objective response rate in participants in the pooled pembrolizumab population by TMB ≥175…

Figure 3
Objective response rate in participants in the pooled pembrolizumab population by TMB ≥175 mut/exome and

Figure 4

Longitudinal outcomes in participants in…

Figure 4

Longitudinal outcomes in participants in the pooled pembrolizumab population by TMB ≥175 mut/exome…

Figure 4
Longitudinal outcomes in participants in the pooled pembrolizumab population by TMB ≥175 mut/exome and

Figure 5

Objective response rate by treatment…

Figure 5

Objective response rate by treatment arm in participants in the 3randomized clinical trials…

Figure 5
Objective response rate by treatment arm in participants in the 3randomized clinical trials included in the analysis by TMB ≥175 mut/exome and
Similar articles
Cited by
References
    1. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science 2015;348:69–74. 10.1126/science.aaa4971 - DOI - PubMed
    1. Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med 2017;377:2500–1. 10.1056/NEJMc1713444 - DOI - PMC - PubMed
    1. Kim JY, Kronbichler A, Eisenhut M, et al. . Tumor mutational burden and efficacy of immune checkpoint inhibitors: a systematic review and meta-analysis. Cancers 2019;11:1798. 10.3390/cancers11111798 - DOI - PMC - PubMed
    1. Osipov A, Lim SJ, Popovic A, et al. . Tumor mutational burden, toxicity, and response of immune checkpoint inhibitors targeting PD(L)1, CTLA-4, and combination: a meta-regression analysis. Clin Cancer Res 2020;26:4842–51. 10.1158/1078-0432.CCR-20-0458 - DOI - PMC - PubMed
    1. Marabelle A, Fakih M, Lopez J, et al. . Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol 2020;21:1353–65. 10.1016/S1470-2045(20)30445-9 - DOI - PubMed
Show all 42 references
Publication types
MeSH terms
Associated data
Related information
Full text links [x]
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 3
Figure 3
Objective response rate in participants in the pooled pembrolizumab population by TMB ≥175 mut/exome and

Figure 4

Longitudinal outcomes in participants in…

Figure 4

Longitudinal outcomes in participants in the pooled pembrolizumab population by TMB ≥175 mut/exome…

Figure 4
Longitudinal outcomes in participants in the pooled pembrolizumab population by TMB ≥175 mut/exome and

Figure 5

Objective response rate by treatment…

Figure 5

Objective response rate by treatment arm in participants in the 3randomized clinical trials…

Figure 5
Objective response rate by treatment arm in participants in the 3randomized clinical trials included in the analysis by TMB ≥175 mut/exome and
Similar articles
Cited by
References
    1. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science 2015;348:69–74. 10.1126/science.aaa4971 - DOI - PubMed
    1. Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med 2017;377:2500–1. 10.1056/NEJMc1713444 - DOI - PMC - PubMed
    1. Kim JY, Kronbichler A, Eisenhut M, et al. . Tumor mutational burden and efficacy of immune checkpoint inhibitors: a systematic review and meta-analysis. Cancers 2019;11:1798. 10.3390/cancers11111798 - DOI - PMC - PubMed
    1. Osipov A, Lim SJ, Popovic A, et al. . Tumor mutational burden, toxicity, and response of immune checkpoint inhibitors targeting PD(L)1, CTLA-4, and combination: a meta-regression analysis. Clin Cancer Res 2020;26:4842–51. 10.1158/1078-0432.CCR-20-0458 - DOI - PMC - PubMed
    1. Marabelle A, Fakih M, Lopez J, et al. . Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol 2020;21:1353–65. 10.1016/S1470-2045(20)30445-9 - DOI - PubMed
Show all 42 references
Publication types
MeSH terms
Associated data
Related information
Full text links [x]
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 4
Figure 4
Longitudinal outcomes in participants in the pooled pembrolizumab population by TMB ≥175 mut/exome and

Figure 5

Objective response rate by treatment…

Figure 5

Objective response rate by treatment arm in participants in the 3randomized clinical trials…

Figure 5
Objective response rate by treatment arm in participants in the 3randomized clinical trials included in the analysis by TMB ≥175 mut/exome and
Similar articles
Cited by
References
    1. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science 2015;348:69–74. 10.1126/science.aaa4971 - DOI - PubMed
    1. Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med 2017;377:2500–1. 10.1056/NEJMc1713444 - DOI - PMC - PubMed
    1. Kim JY, Kronbichler A, Eisenhut M, et al. . Tumor mutational burden and efficacy of immune checkpoint inhibitors: a systematic review and meta-analysis. Cancers 2019;11:1798. 10.3390/cancers11111798 - DOI - PMC - PubMed
    1. Osipov A, Lim SJ, Popovic A, et al. . Tumor mutational burden, toxicity, and response of immune checkpoint inhibitors targeting PD(L)1, CTLA-4, and combination: a meta-regression analysis. Clin Cancer Res 2020;26:4842–51. 10.1158/1078-0432.CCR-20-0458 - DOI - PMC - PubMed
    1. Marabelle A, Fakih M, Lopez J, et al. . Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol 2020;21:1353–65. 10.1016/S1470-2045(20)30445-9 - DOI - PubMed
Show all 42 references
Publication types
MeSH terms
Associated data
Related information
Full text links [x]
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 5
Figure 5
Objective response rate by treatment arm in participants in the 3randomized clinical trials included in the analysis by TMB ≥175 mut/exome and

References

    1. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science 2015;348:69–74. 10.1126/science.aaa4971
    1. Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med 2017;377:2500–1. 10.1056/NEJMc1713444
    1. Kim JY, Kronbichler A, Eisenhut M, et al. . Tumor mutational burden and efficacy of immune checkpoint inhibitors: a systematic review and meta-analysis. Cancers 2019;11:1798. 10.3390/cancers11111798
    1. Osipov A, Lim SJ, Popovic A, et al. . Tumor mutational burden, toxicity, and response of immune checkpoint inhibitors targeting PD(L)1, CTLA-4, and combination: a meta-regression analysis. Clin Cancer Res 2020;26:4842–51. 10.1158/1078-0432.CCR-20-0458
    1. Marabelle A, Fakih M, Lopez J, et al. . Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol 2020;21:1353–65. 10.1016/S1470-2045(20)30445-9
    1. Ribas A, Hamid O, Daud A, et al. . Association of pembrolizumab with tumor response and survival among patients with advanced melanoma. JAMA 2016;315:1600–9. 10.1001/jama.2016.4059
    1. Garon EB, Rizvi NA, Hui R, et al. . Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med 2015;372:2018–28. 10.1056/NEJMoa1501824
    1. Ribas A, Puzanov I, Dummer R, et al. . Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): a randomised, controlled, phase 2 trial. Lancet Oncol 2015;16:908–18. 10.1016/S1470-2045(15)00083-2
    1. Herbst RS, Baas P, Kim D-W, 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. 10.1016/S0140-6736(15)01281-7
    1. Nanda R, Chow LQM, Dees EC, et al. . Pembrolizumab in patients with advanced triple-negative breast cancer: phase Ib KEYNOTE-012 study. J Clin Oncol 2016;34:2460–7. 10.1200/JCO.2015.64.8931
    1. Muro K, Chung HC, Shankaran V, et al. . Pembrolizumab for patients with PD-L1-positive advanced gastric cancer (KEYNOTE-012): a multicentre, open-label, phase 1b trial. Lancet Oncol 2016;17:717–26. 10.1016/S1470-2045(16)00175-3
    1. Plimack ER, Bellmunt J, Gupta S, et al. . Safety and activity of pembrolizumab in patients with locally advanced or metastatic urothelial cancer (KEYNOTE-012): a non-randomised, open-label, phase 1b study. Lancet Oncol 2017;18:212–20. 10.1016/S1470-2045(17)30007-4
    1. Mehra R, Seiwert TY, Gupta S, et al. . Efficacy and safety of pembrolizumab in recurrent/metastatic head and neck squamous cell carcinoma: pooled analyses after long-term follow-up in KEYNOTE-012. Br J Cancer 2018;119:153–9. 10.1038/s41416-018-0131-9
    1. Ott PA, Bang Y-J, Piha-Paul SA, 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. 10.1200/JCO.2018.78.2276
    1. Bellmunt J, de Wit R, Vaughn DJ, et al. . Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N Engl J Med 2017;376:1015–26. 10.1056/NEJMoa1613683
    1. Bauml J, Seiwert TY, Pfister DG, et al. . Pembrolizumab for platinum- and cetuximab-refractory head and neck cancer: results from a single-arm, phase II study. J Clin Oncol 2017;35:1542–9. 10.1200/JCO.2016.70.1524
    1. Fuchs CS, Doi T, Jang RW, et al. . Safety and efficacy of pembrolizumab monotherapy in patients with previously treated advanced gastric and gastroesophageal junction cancer: phase 2 clinical KEYNOTE-059 trial. JAMA Oncol 2018;4:e180013. 10.1001/jamaoncol.2018.0013
    1. Shitara K, Özgüroğlu M, Bang Y-J, et al. . Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): a randomised, open-label, controlled, phase 3 trial. Lancet 2018;392:123–33. 10.1016/S0140-6736(18)31257-1
    1. Adams S, Schmid P, Rugo HS, et al. . Pembrolizumab monotherapy for previously treated metastatic triple-negative breast cancer: cohort A of the phase II KEYNOTE-086 study. Ann Oncol 2019;30:397–404. 10.1093/annonc/mdy517
    1. Matulonis UA, Shapira-Frommer R, Santin AD, et al. . Antitumor activity and safety of pembrolizumab in patients with advanced recurrent ovarian cancer: results from the phase II KEYNOTE-100 study. Ann Oncol 2019;30:1080–7. 10.1093/annonc/mdz135
    1. Antonarakis ES, Piulats JM, Gross-Goupil M, et al. . Pembrolizumab for treatment-refractory metastatic castration-resistant prostate cancer: multicohort, open-label phase II KEYNOTE-199 study. J Clin Oncol 2020;38:395–405. 10.1200/JCO.19.01638
    1. Cristescu R, Mogg R, Ayers M, et al. . Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science 2018;36210.1126/science.aar3593
    1. Ayers M, Lunceford J, Nebozhyn M, et al. . IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest 2017;127:2930–40. 10.1172/JCI91190
    1. Panda A, Betigeri A, Subramanian K, et al. . Identifying a clinically applicable mutational burden threshold as a potential biomarker of response to immune checkpoint therapy in solid tumors. JCO Precis Oncol 2017;2017:1–13. 10.1200/PO.17.00146
    1. Friends of Cancer Research TMB Harmonization Working Group . Tissue agnostic TMB clinical cut-off harmonization initiative. Washington, DC, 2020.
    1. Samstein RM, Lee C-H, Shoushtari AN, et al. . Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet 2019;51:202–6. 10.1038/s41588-018-0312-8
    1. Alexandrov LB, Nik-Zainal S, Wedge DC, et al. . Signatures of mutational processes in human cancer. Nature 2013;500:415–21. 10.1038/nature12477
    1. Castle JC, Uduman M, Pabla S, et al. . Mutation-derived neoantigens for cancer immunotherapy. Front Immunol 2019;10:10. 10.3389/fimmu.2019.01856
    1. Rizvi NA, Hellmann MD, Snyder A, et al. . Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015;348:124–8. 10.1126/science.aaa1348
    1. Chen Y, Liu Q, Chen Z, et al. . PD-L1 expression and tumor mutational burden status for prediction of response to chemotherapy and targeted therapy in non-small cell lung cancer. J Exp Clin Cancer Res 2019;38:193. 10.1186/s13046-019-1192-1
    1. Yu H, Chen Z, Ballman KV, et al. . Correlation of PD-L1 expression with tumor mutation burden and gene signatures for prognosis in early-stage squamous cell lung carcinoma. J Thorac Oncol 2019;14:25–36. 10.1016/j.jtho.2018.09.006
    1. Zhou KI, Peterson B, Serritella A, et al. . Spatial and temporal heterogeneity of PD-L1 expression and tumor mutational burden in gastroesophageal adenocarcinoma at baseline diagnosis and after chemotherapy. Clin Cancer Res 2020;26:6453–63. 10.1158/1078-0432.CCR-20-2085
    1. Özdoğan M, Papadopoulou E, Tsoulos N, et al. . Comprehensive tumor molecular profile analysis in clinical practice. BMC Med Genomics 2021;14:105. 10.1186/s12920-021-00952-9
    1. Hellmann MD, Ciuleanu T-E, Pluzanski A, et al. . Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden. N Engl J Med 2018;378:2093–104. 10.1056/NEJMoa1801946
    1. Chalmers ZR, Connelly CF, Fabrizio D, et al. . Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med 2017;9:34. 10.1186/s13073-017-0424-2
    1. Feliubadaló L, Tonda R, Gausachs M, et al. . Benchmarking of whole exome sequencing and AD hoc designed panels for genetic testing of hereditary cancer. Sci Rep 2017;7:37984. 10.1038/srep37984
    1. Goodman AM, Kato S, Bazhenova L, et al. . Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol Cancer Ther 2017;16:2598–608. 10.1158/1535-7163.MCT-17-0386
    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. Stenzinger A, Allen JD, Maas J, et al. . Tumor mutational burden standardization initiatives: recommendations for consistent tumor mutational burden assessment in clinical samples to guide immunotherapy treatment decisions. Genes Chromosomes Cancer 2019;58:578–88. 10.1002/gcc.22733
    1. Valero C, Lee M, Hoen D, et al. . Response rates to anti-PD-1 immunotherapy in microsatellite-stable solid tumors with 10 or more mutations per Megabase. JAMA Oncol 2021;7:739–43. 10.1001/jamaoncol.2020.7684
    1. McGrail DJ, Pilié PG, Rashid NU, et al. . High tumor mutation burden fails to predict immune checkpoint blockade response across all cancer types. Ann Oncol 2021;32:661–72. 10.1016/j.annonc.2021.02.006
    1. Rousseau B, Foote MB, Maron SB, et al. . The spectrum of benefit from checkpoint blockade in hypermutated tumors. N Engl J Med 2021;384:1168–70. 10.1056/NEJMc2031965

Source: PubMed

3
Se inscrever