Blood-based tumor mutational burden as a biomarker for atezolizumab in non-small cell lung cancer: the phase 2 B-F1RST trial

Edward S Kim, Vamsidhar Velcheti, Tarek Mekhail, Cindy Yun, Sarah M Shagan, Sylvia Hu, Young Kwang Chae, Ticiana A Leal, Jonathan E Dowell, Michaela L Tsai, Christopher S R Dakhil, Philip Stella, Yanling Jin, David S Shames, Erica Schleifman, David A Fabrizio, See Phan, Mark A Socinski, Edward S Kim, Vamsidhar Velcheti, Tarek Mekhail, Cindy Yun, Sarah M Shagan, Sylvia Hu, Young Kwang Chae, Ticiana A Leal, Jonathan E Dowell, Michaela L Tsai, Christopher S R Dakhil, Philip Stella, Yanling Jin, David S Shames, Erica Schleifman, David A Fabrizio, See Phan, Mark A Socinski

Abstract

Tumor mutational burden (TMB) in circulating tumor DNA (ctDNA) has shown promise in predicting benefit from PD-L1/PD-1 inhibitors in retrospective studies. Aiming to assess blood TMB (bTMB) prospectively, we conducted B-F1RST ( NCT02848651 ), an open-label, phase 2 trial that evaluated bTMB as a predictive biomarker for first-line atezolizumab monotherapy in locally advanced or metastatic stage IIIB-IVB non-small cell lung cancer (n = 152). The co-primary endpoints were investigator-assessed objective response rate (ORR) per RECIST version 1.1 and investigator-assessed progression-free survival (PFS) between high and low bTMB subgroups at the pre-defined bTMB ≥ 16 (14.5 mutations per megabase) cutoff. Secondary endpoints included investigator-assessed PFS, overall survival (OS) and duration of response at various bTMB cutoffs, as well as safety. Investigator-assessed PFS in the bTMB ≥ 16 versus bTMB < 16 groups was not statistically significant. However, bTMB ≥ 16 was associated with higher ORR, and ORR improved as bTMB cutoffs increased. No new safety signals were seen. In exploratory analyses, patients with maximum somatic allele frequency (MSAF) < 1% had higher ORR than patients with MSAF ≥ 1%. However, further analysis showed that this effect was driven by better baseline prognostics rather than by MSAF itself. At 36.5-month follow-up, an exploratory analysis of OS found that bTMB ≥ 16 was associated with longer OS than bTMB < 16. Further study and assay optimization will be required to develop bTMB as a predictive, standalone biomarker of immunotherapy or for use in conjunction with other biomarkers.

Conflict of interest statement

The authors declare the following competing interests: E.S.K. has received consulting fees from AstraZeneca, Merck and Roche/Genentech. V.V. has received fees for consulting or serving on advisory boards for Bristol Myers Squibb, Merck, GlaxoSmithKline, Foundation Medicine, AstraZeneca, EMD Serono, Novartis and Novocure. T.M declares no competing interests. C.Y. is an employee of Genentech and holds stock in Roche. S.M.S. is an employee of Genentech and holds stock in Roche. S.H. is employed by Genentech and holds stock in Roche. Y.K.C. has received research grants from AbbVie, Bristol Myers Squibb, Biodesix, Lexent Bio and Freenome and fees for serving on advisory boards for Roche/Genentech, Bristol Myers Squibb, AstraZeneca, Merck, Foundation Medicine, Counsyl, Neogenomics, Guardant Health, Boehringher Ingelheim, Biodesix, ImmuneOncia, Lilly Oncology, Merck, Takeda, Pfizer, Tempus, Lunit and Jazz Pharmaceuticals. T.A.L. has received fees for serving on advisory boards for Jazz Pharmaceuticals, AstraZeneca, EMD Serono, Merck, Blueprint, Debio and Bayer and consultancy for Boehringer Ingelheim, Daiichi Sankyo, Genentech and Jazz Pharmaceuticals. T.A.L. has also served as a non-compensated member of the steering committee for the SAPPHIRE trial, sponsored by Mirati Therapeutics. J.E.D. has received fess for serving on advisory boards for AstraZeneca, Genentech and Janssen. M.L.T., S.R.D. and P.S. declare no competing interests. Y.J. is an employee of and holds stock in Roche. D.S.S. is an employee of Genentech and holds stock in Roche. E.S. is an employee of Genentech and holds stock in Roche. D.A.F. is an employee of Foundation Medicine and holds stock in Roche. S.P. is an employee of Genentech and holds stock in Roche. M.A.S. has received research grants from Genentech, Spectrum, AstraZeneca, Novartis and Daichii Sankyo and has received fees for serving on speaker bureaus for Genentech, AstraZeneca, Bayer, Novartis, Guardant and Amgen.

© 2022. The Author(s).

Figures

Fig. 1. Confirmed responses with atezolizumab.
Fig. 1. Confirmed responses with atezolizumab.
ORRs are shown for bTMB-high (dark blue) and bTMB-low (light blue) subgroups at three different cutoffs, all of which showed significant differences between the subgroups. Data cut: 26 July 2019. Statistical tests (Cochran–Mantel–Haenszel) were unadjusted for multiple comparisons and two-sided at the 0.10 significance level. Rate difference (90% CI): bTMB ≥ 10, 13.3% (2.5, 24.0); bTMB ≥ 16, 30.2% (14.8, 45.6); and bTMB ≥ 20, 41.4% (22.1, 60.6). CR, complete response; PR, partial response.
Fig. 2. Final analysis outcomes at the…
Fig. 2. Final analysis outcomes at the bTMB16 cutoff.
a. Kaplan–Meier survival curves of PFS for bTMB < 16 (light blue) versus bTMB ≥ 16 (dark blue). b. Kaplan–Meier survival curves of OS for bTMB < 16 (light blue) versus bTMB ≥ 16 (dark blue). Data cut: 26 July 2019. Statistical tests (log-rank) were unadjusted for multiple comparisons and two-sided at the 0.10 significance level. m, median; mo, months; NE, not estimable.
Fig. 3. Depth of response according to…
Fig. 3. Depth of response according to PD-L1 and bTMB categories.
This waterfall plot shows the maximum change in SLD from baseline by response category. PD-L1a and bTMB status by category are displayed below the graph. Patients without a post-baseline SLD assessment were not included. Data cut: 26 July 2019. N/A, not available; PD, progressive disease; PR, partial response; SD, stable disease. a PD-L1 status was determined by any standard-of-care assay.
Fig. 4. PFS and OS HRs at…
Fig. 4. PFS and OS HRs at different bTMB cutoffs.
a, PFS by bTMB cutoff. b, OS by bTMB cutoff. n refers to the number of patients in the respective subgroup. The blue diamonds indicate the HR. Statistical tests (log-rank) were unadjusted for multiple comparisons and two-sided at the 0.10 significance level. The error bars show the 90% CI. mo, months.
Fig. 5. Long-term follow-up OS. Kaplan–Meier plots…
Fig. 5. Long-term follow-up OS. Kaplan–Meier plots of OS at the bTMB ≥ 16 cutoff.
Statistical tests (log-rank) were unadjusted for multiple comparisons and two-sided at the 0.10 significance level. m, median; mo, months; NE, not estimable.
Fig. 6. Atezolizumab AEs observed in ≥…
Fig. 6. Atezolizumab AEs observed in ≥ 10% of the ITT population.
The incidence of AEs by grade ≥ 3 (dark blue) and grade 1–2 (light blue).
Extended Data Fig. 1. B-F1RST study design.
Extended Data Fig. 1. B-F1RST study design.
B-F1RST prospectively evaluated atezolizumab monotherapy in the first-line treatment of patients with NSCLC. bTMB, blood-based tumor mutational burden; DOR, duration of response; ECOG PS, Eastern Cooperative Oncology Group performance status; INV, investigator; NSCLC, non-small cell lung cancer; OS, overall survival; PD, progressive disease; PD-L1, programmed death-ligand 1; PFS, progression-free survival; RECIST v1.1, Response Evaluation Criteria in Solid Tumors version 1.1. a Staging based on International Association for the Study of Lung Cancer Lung Cancer Staging Project 8th Edition of the TNM Classification for Lung Cancer. b Total enrolled, N = 153; however, 1 patient was never treated and was not included in the intention-to-treat population. c Tissue biopsy was optional.
Extended Data Fig. 2. Patient flow.
Extended Data Fig. 2. Patient flow.
153 patients were allocated to receive atezolizumab. One patient was not treated and was excluded from the primary analysis. The biomarker-evaluable population excluded patients with MSAF a Staging based on International Association for the Study of Lung Cancer Lung Cancer Staging Project 8th Edition of the TNM Classification for Lung Cancer.
Extended Data Fig. 3. Unadjusted and adjusted…
Extended Data Fig. 3. Unadjusted and adjusted MSAF-associated PFS.
a. Kaplan–Meier plots of unadjusted PFS. b. Kaplan–Meier plot of PFS adjusted for baseline imbalances. After the PFS curves were adjusted, the difference between arms was not significant. Statistical tests (log-rank) were unadjusted for multiple comparisons and 2-sided at the 0.05 significance level. HR, hazard ratio; m, median; MSAF, maximum somatic allele frequency; PFS, progression-free survival.
Extended Data Fig. 4. Mutant genes in…
Extended Data Fig. 4. Mutant genes in bTMB subgroups with > 2% frequency.
a. The prevalence of genes in the MEP, MSAF ≥ 1%, bTMB ≥ 16 and bTMB < 16 populations. The bTMB-high population was enriched in TP53 mutations compared with the other groups. Differences for all other genes were not significant. bTMB, blood-based tumor mutational burden; MEP, molecularly evaluable population; MSAF, maximum somatic allele frequency. aDoes not include FANCD2.

References

    1. Herbst RS, et al. Atezolizumab for first-line treatment of PD-L1-selected patients with NSCLC. N. Engl. J. Med. 2020;383:1328–1339. doi: 10.1056/NEJMoa1917346.
    1. Carbone DP, et al. First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N. Engl. J. Med. 2017;376:2415–2426. doi: 10.1056/NEJMoa1613493.
    1. Kowanetz M, et al. Abstract OA20.01: Tumor mutation burden (TMB) is associated with improved efficacy of atezolizumab in 1L and 2L+ NSCLC patients. J. Thorac. Oncol. 2017;12:S321–S322. doi: 10.1016/j.jtho.2016.11.343.
    1. Herbst RS, et al. Association between tissue TMB (tTMB) and clinical outcomes with pembrolizumab monotherapy (pembro) in PD-L1-positive advanced NSCLC in the KEYNOTE-010 and -042 trials. Ann. Oncol. 2019;30:v851–v934. doi: 10.1093/annonc/mdy545.
    1. Peters S, et al. Abstract CT074: Tumor mutational burden (TMB) as a biomarker of survival in metastatic non-small cell lung cancer (mNSCLC): blood and tissue TMB analysis from MYSTIC, a phase III study of first-line durvalumab ± tremelimumab vs chemotherapy. Cancer Res. 2019;79:CT074–CT074.
    1. Borghaei H, et al. Nivolumab (Nivo) + platinum-doublet chemotherapy (Chemo) vs chemo as first-line (1L) treatment (Tx) for advanced non-small cell lung cancer (NSCLC) with <1% tumor PD-L1 expression: results from CheckMate 227. J. Clin. Oncol. 2018;36:9001–9001. doi: 10.1200/JCO.2018.36.15_suppl.9001.
    1. Lim C, et al. Biomarker testing and time to treatment decision in patients with advanced nonsmall-cell lung cancer. Ann. Oncol. 2015;26:1415–1421. doi: 10.1093/annonc/mdv208.
    1. Jamal-Hanjani M, et al. Tracking the evolution of non-small-cell lung cancer. N. Engl. J. Med. 2017;376:2109–2121. doi: 10.1056/NEJMoa1616288.
    1. Cai W, et al. Intratumoral heterogeneity of ALK-rearranged and ALK/EGFR coaltered lung adenocarcinoma. J. Clin. Oncol. 2015;33:3701–3709. doi: 10.1200/JCO.2014.58.8293.
    1. Gandara DR, et al. Blood-based tumor mutational burden as a predictor of clinical benefit in non-small-cell lung cancer patients treated with atezolizumab. Nat. Med. 2018;24:1441–1448. doi: 10.1038/s41591-018-0134-3.
    1. Kim ES, et al. Abstract 4855: Primary efficacy results from B-F1RST, a prospective phase II trial evaluating blood-based tumour mutational burden (bTMB) as a predictive biomarker for atezolizumab (atezo) in 1L non-small cell lung cancer (NSCLC) Ann. Oncol. 2018;29:viii744. doi: 10.1093/annonc/mdy424.067.
    1. Socinski M, et al. Abstract CT194: Exploratory subgroup analysis of atezolizumab (atezo) clinical characteristics in patients (pts) with low circulating tumor DNA (ctDNA) in B-F1RST—a phase II trial evaluating blood-based tumor mutational burden (bTMB) in NSCLC. Cancer Res. 2019;79:CT194.
    1. Motta G, et al. Considerations about tumor size as a factor of prognosis in NSCLC. Ann. Ital. Chir. 1999;70:893–897.
    1. Hirsch FR, et al. The prognostic and predictive role of histology in advanced non-small cell lung cancer: a literature review. J. Thorac. Oncol. 2008;3:1468–1481. doi: 10.1097/JTO.0b013e318189f551.
    1. Cuyún Carter G, et al. A comprehensive review of nongenetic prognostic and predictive factors influencing the heterogeneity of outcomes in advanced non-small-cell lung cancer. Cancer Manag. Res. 2014;6:437–449. doi: 10.2147/CMAR.S63603.
    1. Hellmann MD, et al. Nivolumab plus ipilimumab in advanced non-small-cell lung cancer. N. Engl. J. Med. 2019;381:2020–2031. doi: 10.1056/NEJMoa1910231.
    1. Rizvi NA, et al. Abstract LBA6: Durvalumab with or without tremelimumab vs platinum-based chemotherapy as first-line treatment for metastatic non-small cell lung cancer: MYSTIC. Ann. Oncol. 2018;29:mdy511.005–mdy511.005. doi: 10.1093/annonc/mdy511.005.
    1. Dziadziuszko R, et al. Abstract 1281O: Atezolizumab vs platinum-based chemotherapy in blood-based tumour mutational burden–positive NSCLC: results of the blood first assay screening trial (BFAST) phase 3 Cohort C. Ann. Oncol. 2021;32:S950–S951. doi: 10.1016/j.annonc.2021.08.1883.
    1. Sholl LM, 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;15:1409–1424. doi: 10.1016/j.jtho.2020.05.019.
    1. Keytruda (pembrolizumab) [prescribing information] (2020).
    1. Turajlic S, et al. Insertion-and-deletion-derived tumour-specific neoantigens and the immunogenic phenotype: a pan-cancer analysis. Lancet Oncol. 2017;18:1009–1021. doi: 10.1016/S1470-2045(17)30516-8.
    1. Jardim DL, Goodman A, de Melo Gagliato D, Kurzrock R. The challenges of tumor mutational burden as an immunotherapy biomarker. Cancer Cell. 2021;39:154–173. doi: 10.1016/j.ccell.2020.10.001.
    1. Goodman AM, et al. MHC-I genotype and tumor mutational burden predict response to immunotherapy. Genome Med. 2020;12:45. doi: 10.1186/s13073-020-00743-4.
    1. Nabet BY, et al. Noninvasive early identification of therapeutic benefit from immune checkpoint inhibition. Cell. 2020;183:363–376. doi: 10.1016/j.cell.2020.09.001.
    1. Rizvi H, et al. Molecular determinants of response to anti-programmed cell death (PD)-1 and anti-programmed death-ligand 1 (PD-L1) blockade in patients with non-small-cell lung cancer profiled with targeted next-generation sequencing. J. Clin. Oncol. 2018;36:633–641. doi: 10.1200/JCO.2017.75.3384.
    1. Goldberg SB, et al. Pembrolizumab for patients with melanoma or non-small-cell lung cancer and untreated brain metastases: early analysis of a non-randomised, open-label, phase 2 trial. Lancet Oncol. 2016;17:976–983. doi: 10.1016/S1470-2045(16)30053-5.
    1. Raja R, et al. Early reduction in ctDNA predicts survival in patients with lung and bladder cancer treated with durvalumab. Clin. Cancer Res. 2018;24:6212–6222. doi: 10.1158/1078-0432.CCR-18-0386.
    1. Goldstraw P, et al. The IASLC Lung Cancer Staging Project: proposals for revision of the TNM stage groupings in the forthcoming (8th) edition of the TNM Classification for Lung Cancer. J. Thorac. Onc. 2015;11:39–51. doi: 10.1016/j.jtho.2015.09.009.

Source: PubMed

3
구독하다