Phase 2 study of pembrolizumab in patients with recurrent and residual high-grade meningiomas

Priscilla K Brastianos, Albert E Kim, Anita Giobbie-Hurder, Eudocia Quant Lee, Nancy Wang, April F Eichler, Ugonma Chukwueke, Deborah A Forst, Isabel C Arrillaga-Romany, Jorg Dietrich, Zachary Corbin, Jennifer Moliterno, Joachim Baehring, Michael White, Kevin W Lou, Juliana Larson, Magali A de Sauvage, Kathryn Evancic, Joana Mora, Naema Nayyar, Jay Loeffler, Kevin Oh, Helen A Shih, William T Curry, Daniel P Cahill, Fred G Barker, Elizabeth R Gerstner, Sandro Santagata, Priscilla K Brastianos, Albert E Kim, Anita Giobbie-Hurder, Eudocia Quant Lee, Nancy Wang, April F Eichler, Ugonma Chukwueke, Deborah A Forst, Isabel C Arrillaga-Romany, Jorg Dietrich, Zachary Corbin, Jennifer Moliterno, Joachim Baehring, Michael White, Kevin W Lou, Juliana Larson, Magali A de Sauvage, Kathryn Evancic, Joana Mora, Naema Nayyar, Jay Loeffler, Kevin Oh, Helen A Shih, William T Curry, Daniel P Cahill, Fred G Barker, Elizabeth R Gerstner, Sandro Santagata

Abstract

High-grade meningiomas are associated with neuro-cognitive morbidity and have limited treatments. High-grade meningiomas harbor an immunosuppressive tumor microenvironment (TME) and programmed death-ligand 1 (PD-L1) expression may contribute to their aggressive phenotype. Here, we present the results of a single-arm, open-label phase 2 trial (NCT03279692) evaluating the efficacy of pembrolizumab, a PD-1 inhibitor, in a cohort of 25 evaluable patients with recurrent and progressive grade 2 and 3 meningiomas. The primary endpoint is the proportion of patients alive and progression-free at 6 months (PFS-6). Secondary endpoints include progression-free and overall survival, best intracranial response, and toxicity. Our study has met its primary endpoint and achieved a PFS-6 rate of 0.48 (90% exact CI: 0.31-0.66) and a median PFS of 7.6 months (90% CI: 3.4-12.9 months). Twenty percent of patients have experienced one (or more) grade-3 or higher treatment-related adverse events. These results suggest that pembrolizumab exerts promising efficacy on a subset of these tumors. Further studies are needed to identify the biological facets within the meningioma TME that may drive response to immune-based therapies.

Conflict of interest statement

PKB has consulted for Tesaro, Angiochem, Genentech-Roche, ElevateBio, Eli Lilly, SK Life Sciences, Advise Connect Inspire (ACI), Pfizer, Voyager Therapeutics, Sintetica, and Dantari, received institutional research funding (to MGH) from Merck, Mirati, Eli Lilly, BMS, and Pfizer, and has received honoraria from Merck, Pfizer, and Genentech-Roche. DAF has Eli Lilly stock ownership. ICA has received institutional research support from Astex Pharmaceuticals and consulted for Boehringer Ingelheim, FORMA Therapeutics, and Agios. DPC has consulted for Lilly, GlaxoSmithKline, Boston Pharmaceuticals, and Iconovir, and serves on the advisory board of Pyramid Biosciences, which includes an equity interest, and has received honoraria and travel reimbursement from Merck for invited lectures, and funding from the US NIH and DOD for clinical trial and grant review. Finally, pembrolizumab is a drug that was developed based in part on scientific research from Dana-Farber Cancer Institute (DFCI), which was licensed to Merck US, the pharmaceutical company that manufactures and sells pembrolizumab. Through the licensing arrangement, DFCI may receive money from this company. Any research that might affect the sales or use of pembrolizumab could change the payments DFCI receives from this company. This is known as an institutional conflict of interest. The remaining authors declare no competing interests.

© 2022. The Author(s).

Figures

Fig. 1. Kaplan–Meier curve for progression-free survival.
Fig. 1. Kaplan–Meier curve for progression-free survival.
Kaplan–Meier estimates of PFS are shown. There were 22 PFS events among 25 patients. The median PFS was 7.6 months (90% CI: 3.4–12.9).
Fig. 2. Clinical course of high-grade meningioma…
Fig. 2. Clinical course of high-grade meningioma patients treated with pembrolizumab.
Time of best radiographic response (as defined by TIMC), death, and study-limiting toxicity for the patient population. The arrow at the end of the bar indicates the time at which the patient was last known to be alive at the time of data analysis. The dashed line at 6 months is the threshold of the primary endpoint (PFS-6). Patients marked in blue had extracranial or metastatic meningiomas. One patient (MEN_06) was excluded from this schematic, as they withdrew consent one week after enrollment before receiving pembrolizumab.

References

    1. Ostrom QT, et al. CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2013–2017. Neuro. Oncol. 2020;22:IV1–IV96. doi: 10.1093/neuonc/noaa200.
    1. Buerki RA, et al. An overview of meningiomas. Future Oncol. 2018;14:2161–2177. doi: 10.2217/fon-2018-0006.
    1. Kaley T, et al. Historical benchmarks for medical therapy trials in surgery-and radiation-refractory meningioma: a RANO review. Neuro. Oncol. 2014;16:829–840. doi: 10.1093/neuonc/not330.
    1. Bi WL, et al. Genomic landscape of high-grade meningiomas. NPJ Genom. Med. 2017;2:15. doi: 10.1038/s41525-017-0014-7.
    1. Brastianos PK, et al. Genomic sequencing of meningiomas identifies oncogenic SMO and AKT1 mutations. Nat. Genet. 2013;45:285–289. doi: 10.1038/ng.2526.
    1. Karimi, S. et al. Programmed death ligand-1 (PD-L1) expression in meningioma; prognostic significance and its association with hypoxia and NFKB2 expression. Sci. Rep. 10, 14115 (2020).
    1. Everson RG, et al. Multiplatform profiling of meningioma provides molecular insight and prioritization of drug targets for rational clinical trial design. J. Neurooncol. 2018;139:469–478. doi: 10.1007/s11060-018-2891-8.
    1. Yeung J, et al. Spatially resolved and quantitative analysis of the immunological landscape in human meningiomas. J. Neuropathol. Exp. Neurol. 2021;80:150–159. doi: 10.1093/jnen/nlaa152.
    1. Dunn, I. F. et al. Mismatch repair deficiency in high-grade meningioma: a rare but recurrent event associated with dramatic immune activation and clinical response to PD-1 blockade. JCO Precis. Oncol., 10.1200/po.18.00190 (2018).
    1. Han SJ, et al. Expression and prognostic impact of immune modulatory molecule PD-L1 in meningioma. J. Neurooncol. 2016;130:543–552. doi: 10.1007/s11060-016-2256-0.
    1. Blume, C. et al. Integrated phospho-proteogenomic and single-cell transcriptomic analysis of meningiomas establishes robust subtyping and reveals subtype-specific immune invasion. 10.1101/2021.05.11.443369 (2021).
    1. J., Yeung et al. OUP accepted manuscript. Neuro. Oncol., 10.1093/neuonc/noab075 (2021).
    1. Nebot-Bral L, et al. Hypermutated tumours in the era of immunotherapy: the paradigm of personalised medicine. Eur. J. Cancer. 2017;84:290–303. doi: 10.1016/j.ejca.2017.07.026.
    1. Du Z, et al. Increased expression of the immune modulatory molecule PDL1 (CD274) in anaplastic meningioma. Oncotarget. 2015;6:4704–4716. doi: 10.18632/oncotarget.3082.
    1. Powles T, et al. Pembrolizumab plus axitinib versus sunitinib monotherapy as first-line treatment of advanced renal cell carcinoma (KEYNOTE-426): extended follow-up from a randomised, open-label, phase 3 trial. Lancet Oncol. 2020;21:1563–1573. doi: 10.1016/S1470-2045(20)30436-8.
    1. Mok TSK, et al. Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): a randomised, open-label, controlled, phase 3 trial. Lancet. 2019;393:1819–1830. doi: 10.1016/S0140-6736(18)32409-7.
    1. Goldberg SB, et al. Pembrolizumab for management of patients with NSCLC and brain metastases: long-term results and biomarker analysis from a non-randomised, open-label, phase 2 trial. Lancet Oncol. 2020;21:655–663. doi: 10.1016/S1470-2045(20)30111-X.
    1. Hamid O, et al. Five-year survival outcomes for patients with advanced melanoma treated with pembrolizumab in KEYNOTE-001. Ann. Oncol. 2019;30:582–588. doi: 10.1093/annonc/mdz011.
    1. Dalle Ore CL, et al. Meningioma metastases: incidence and proposed screening paradigm. J. Neurosurg. 2019;132:1447–1455. doi: 10.3171/2019.1.JNS181771.
    1. Surov A, et al. Diffusion-weighted imaging in meningioma: Prediction of tumor grade and association with histopathological parameters. Transl. Oncol. 2015;8:517–523. doi: 10.1016/j.tranon.2015.11.012.
    1. Larkin J, et al. Five-year survival with combined Nivolumab and Ipilimumab in advanced melanoma. N. Engl. J. Med. 2019;381:1535–1546. doi: 10.1056/NEJMoa1910836.
    1. Chiou VL, Burotto M. Pseudoprogression and immune-related response in solid tumors. J. Clin. Oncol. 2015;33:3541. doi: 10.1200/JCO.2015.61.6870.
    1. Park, H. J. et al. Incidence of pseudoprogression during immune checkpoint inhibitor therapy for solid tumors: a systematic review and meta-analysis. Radiology. 297, 87–96 (2020).
    1. Emens LA, Middleton G. The interplay of immunotherapy and chemotherapy: harnessing potential synergies. Cancer Immunol. Res. 2015;3:436. doi: 10.1158/2326-6066.CIR-15-0064.
    1. Ellingson BM, Wen PY, Cloughesy TF. Modified criteria for radiographic response assessment in glioblastoma clinical trials. Neurotherapeutics. 2017;14:307–320. doi: 10.1007/s13311-016-0507-6.
    1. Eisenhauer, E. A. et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur. J. Cancer, 10.1016/j.ejca.2008.10.026 (2009).
    1. Dolled-Filhart M, et al. Development of a prototype immunohistochemistry assay to measure programmed death ligand-1 expression in tumor tissue. Arch. Pathol. Lab. Med. 2016;140:1259–1266. doi: 10.5858/arpa.2015-0544-OA.
    1. Garon EB, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N. Engl. J. Med. 2015;372:2018–2028. doi: 10.1056/NEJMoa1501824.
    1. Muro K, et al. Pembrolizumab for patients with PD-L1-positive advanced gastric cancer (KEYNOTE-012): a multicentre, open-label, phase 1b trial. Lancet Oncol. 2016;17:717–726. doi: 10.1016/S1470-2045(16)00175-3.
    1. Nanda, R. et al. Pembrolizumab in patients with advanced triple-negative breast cancer: phase Ib KEYNOTE-012 study. J. Clin. Oncol. 34, 2460–2467 (2016).
    1. Adams S. Dramatic response of metaplastic breast cancer to chemo-immunotherapy. NPJ Breast Cancer. 2017;3:8. doi: 10.1038/s41523-017-0011-0.
    1. R., Cristescu et al. Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science362, eaar3593 (2018).
    1. Joseph RW, et al. Baseline tumor size is an independent prognostic factor for overall survival in patients with melanoma treated with pembrolizumab. Clin. Cancer Res. 2018;24:4960–4967. doi: 10.1158/1078-0432.CCR-17-2386.
    1. Clouthier DL, et al. An interim report on the investigator-initiated phase 2 study of pembrolizumab immunological response evaluation (INSPIRE) J. Immunother. Cancer. 2019;7:1–12. doi: 10.1186/s40425-019-0541-0.
    1. Mehnert JM, et al. Safety and antitumor activity of the anti–PD-1 antibody pembrolizumab in patients with advanced, PD-L1–positive papillary or follicular thyroid cancer. BMC Cancer. 2019;19:1–9. doi: 10.1186/s12885-019-5380-3.
    1. Habra, M. A. et al. Phase II clinical trial of pembrolizumab efficacy and safety in advanced adrenocortical carcinoma. J. Immunother. Cancer. 7, 253 (2019).
    1. Rodriguez CP, et al. A phase II trial of pembrolizumab and vorinostat in recurrent metastatic head and neck squamous cell carcinomas and salivary gland cancer. Clin. Cancer Res. 2020;26:837–845. doi: 10.1158/1078-0432.CCR-19-2214.
    1. Chung HC, et al. Pembrolizumab after two or more lines of previous therapy in patients with recurrent or metastatic SCLC: results from the KEYNOTE-028 and KEYNOTE-158 studies. J. Thorac. Oncol. 2020;15:618–627. doi: 10.1016/j.jtho.2019.12.109.
    1. Hendry S, et al. Assessing tumor-infiltrating lymphocytes in solid tumors: a practical review for pathologists and proposal for a standardized method from the International Immuno-Oncology Biomarkers Working Group: Part 2: TILs in melanoma, gastrointestinal tract carcinomas, non-small cell lung carcinoma and mesothelioma, endometrial and ovarian carcinomas, squamous cell carcinoma of the head and neck, genitourinary carcinomas, and primary brain tumors. Adv. Anat. Pathol. 2017;24:311–335. doi: 10.1097/PAP.0000000000000161.
    1. Beers, A. et al. DeepNeuro: an open-source deep learning toolbox for neuroimaging. Neuroinformatics, 10.1007/s12021-020-09477-5 (2020).

Source: PubMed

3
订阅