Phase I study of a novel glioblastoma radiation therapy schedule exploiting cell-state plasticity

Jamie A Dean, Shyam K Tanguturi, Daniel Cagney, Kee-Young Shin, Gilbert Youssef, Ayal Aizer, Rifaquat Rahman, Lubna Hammoudeh, David Reardon, Eudocia Lee, Jorg Dietrich, Kaoru Tamura, Masaru Aoyagi, Lacey Wickersham, Patrick Y Wen, Paul Catalano, Daphne Haas-Kogan, Brian M Alexander, Franziska Michor, Jamie A Dean, Shyam K Tanguturi, Daniel Cagney, Kee-Young Shin, Gilbert Youssef, Ayal Aizer, Rifaquat Rahman, Lubna Hammoudeh, David Reardon, Eudocia Lee, Jorg Dietrich, Kaoru Tamura, Masaru Aoyagi, Lacey Wickersham, Patrick Y Wen, Paul Catalano, Daphne Haas-Kogan, Brian M Alexander, Franziska Michor

Abstract

Background: Glioblastomas comprise heterogeneous cell populations with dynamic, bidirectional plasticity between treatment-resistant stem-like and treatment-sensitive differentiated states, with treatment influencing this process. However, current treatment protocols do not account for this plasticity. Previously, we generated a mathematical model based on preclinical experiments to describe this process and optimize a radiation therapy fractionation schedule that substantially increased survival relative to standard fractionation in a murine glioblastoma model.

Methods: We developed statistical models to predict the survival benefit of interventions to glioblastoma patients based on the corresponding survival benefit in the mouse model used in our preclinical study. We applied our mathematical model of glioblastoma radiation response to optimize a radiation therapy fractionation schedule for patients undergoing re-irradiation for glioblastoma and developed a first-in-human trial (NCT03557372) to assess the feasibility and safety of administering our schedule.

Results: Our statistical modeling predicted that the hazard ratio when comparing our novel radiation schedule with a standard schedule would be 0.74. Our mathematical modeling suggested that a practical, near-optimal schedule for re-irradiation of recurrent glioblastoma patients was 3.96 Gy × 7 (1 fraction/day) followed by 1.0 Gy × 9 (3 fractions/day). Our optimized schedule was successfully administered to 14/14 (100%) patients.

Conclusions: A novel radiation therapy schedule based on mathematical modeling of cell-state plasticity is feasible and safe to administer to glioblastoma patients.

Keywords: cell-state plasticity; clinical trial; glioblastoma; mathematical modeling; radiation oncology.

Conflict of interest statement

B.A. is an employee of Foundation Medicine and Roche. P.Y.W. receives research support from Astra Zeneca/Medimmune, Beigene, Black Diamond, Celgene, Chimerix, Eli Lily, ERASCA, Genentech/Roche, Kazia, MediciNova, Merck, Novartis, Nuvation Bio, Puma, Servier, Vascular Biogenics, and VBI Vaccines, and serves on the advisory board of Astra Zeneca, Bayer, Black Diamond, Boehringer Ingelheim, Boston Pharmaceuticals, Celularity, Chimerix, Day One Bio, Genenta, Glaxo Smith Kline, Karyopharm, Merck, Mundipharma, Novartis, Novocure, Nuvation Bio, Prelude Therapeutics, Sapience, Servier, Sagimet, Vascular Biogenics, VBI Vaccines. A.A. receives research funding from Varian and NH TherAGuIX and is consulting for Novartis and Seagen. F.M. is a cofounder of and has equity in Harbinger Health, has equity in Zephyr AI, and serves as a consultant for Harbinger Health, Zephyr AI, and Red Cell Partners. D.H.-K. is on the scientific Advisory Board of EmpNia, Inc., and Graegis’s Scientific. JD received research support from Novartis and is a consultant for Unum Therapeutics and Amgen. The authors declare that none of these relationships are directly or indirectly related to the content of this manuscript.

© The Author(s) 2022. Published by Oxford University Press on behalf of the Society for Neuro-Oncology.

Figures

Figure 1.
Figure 1.
Mathematical modeling identifies a radiation therapy administration schedule predicted to improve survival of recurrent GBM patients. (A) Predicted tumor volume dynamics resulting from a radiation schedule of 51.6 Gy in 30 fractions with three times daily dosing with 3.25 h interfraction intervals compared to those from a schedule of 35 Gy in 10 fractions. (B) Predicted stem-like cell dynamics resulting from the administration schedules in (A). (C) Predicted effect of different two-phase radiation administration schedules on tumor volume dynamics. (D) Predicted stem-like cell dynamics resulting from the administration schedules in (C). QD—once daily dosing; TID—three times daily dosing.
Figure 2.
Figure 2.
Patterns of tumor response and survival of patients treated in the clinical trial. (A) Kaplan-Meier plot for progression-free survival. (B) Kaplan-Meier plot for overall survival. (C) Cox proportional hazards regression model hazard ratios for progression-free survival. (D) Cox proportional hazards regression model hazard ratios for overall survival. (E) Swimmer plot of tumor response. The points indicate the responses at individual time points and the colors indicate the best response for each patient. (F) MRI scans of Patient 6 at the time of radiation therapy planning (left), 3 month follow-up (center) and 6 month follow-up with progression (right) showing a representative example of stable to regressed disease locally within the Planning Target Volume (contour line) but evidence of progressive enhancing, hypercellular disease with elevated cerebral blood volume immediately anterior to the Planning Target Volume. The patient died of disease 5 weeks after this time point. (G) MRI scans of Patient 1 at the time of radiotherapy planning (left) and 1 month follow-up (right) showing an example of a distant progression outside of the radiation therapy field (arrow), with stable to well controlled disease locally within the Planning Target Volume (contour line).

References

    1. Wen PY, Weller M, Lee EQ, et al. . Glioblastoma in adults: a Society for Neuro-Oncology (SNO) and European Society of Neuro-Oncology (EANO) consensus review on current management and future directions. Neuro Oncol. 2020;22(8):1073–1113.
    1. Walker MD, Green SB, Byar DP, et al. . Randomized comparisons of radiotherapy and nitrosoureas for the treatment of malignant glioma after surgery. N Engl J Med. 1980;303(23):1323–1329.
    1. Stupp R, Mason WP, van den Bent MJ, et al. ; European Organisation for Research and Treatment of Cancer Brain Tumor and Radiotherapy Groups. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987–996.
    1. Herrlinger U, Tzaridis T, Mack F, et al. . Lomustine-temozolomide combination therapy versus standard temozolomide therapy in patients with newly diagnosed glioblastoma with methylated MGMT promoter (CeTeG/NOA–09): a randomised, open-label, phase 3 trial. Lancet. 2019;393(10172):678–688.
    1. Stupp R, Taillibert S, Kanner A, et al. . Effect of tumor-treating fields plus maintenance temozolomide vs maintenance temozolomide alone on survival in patients with glioblastoma a randomized clinical trial. JAMA. 2017;318(23):2306–2316.
    1. Fowler JF. The linear-quadratic formula and progress in fractionated radiotherapy. Br J Radiol. 1989;62(740):679–694.
    1. Gupta T, Dinshaw K. Modified optimal fractionation for poor prognosis malignant gliomas: An elusive search. Acta Oncol. 2005;44(2):105–113.
    1. Patel AP, Tirosh I, Trombetta JJ, et al. . Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science. 2014;344(6190):1396–1401.
    1. Suvà ML, Rheinbay E, Gillespie SM, et al. . Reconstructing and reprogramming the tumor-propagating potential of glioblastoma stem-like cells. Cell. 2014;157(3):580–594.
    1. Charles N, Ozawa T, Squatrito M, et al. . Perivascular nitric oxide activates notch signaling and promotes stem-like character in PDGF-induced glioma cells. Cell Stem Cell. 2010;6(2):141–152.
    1. Bao S, Wu Q, McLendon RE, et al. . Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006;444(7120):756–760.
    1. Hambardzumyan D, Amankulor NM, Helmy KY, Becher OJ, Holland EC. Modeling adult gliomas using RCAS/t-va technology. Transl Oncol. 2009;2(2):89–95.
    1. Leder K, Pitter K, Laplant Q, et al. . Mathematical modeling of PDGF-driven glioblastoma reveals optimized radiation dosing schedules. Cell. 2014;156(3):603–616.
    1. Mayer R, Sminia P. Reirradiation tolerance of the human brain. Int J Radiat Oncol Biol Phys. 2008;70(5):1350–1360.
    1. Beauchesne P, Bernier V, Carnin C, et al. . Prolonged survival for patients with newly diagnosed, inoperable glioblastoma with 3-times daily ultrafractionated radiation therapy. Neuro Oncol. 2010;12(6):595–602.
    1. Wen PY, Macdonald DR, Reardon DA, et al. . Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol. 2010;28(11):1963–1972.
    1. Friedrich S, Friede T. Causal inference methods for small non-randomized studies: methods and an application in COVID-19. Contemp Clin Trials. 2020;99(January):106213.
    1. Cirkel GA, Weeber F, Bins S, et al. . The time to progression ratio: a new individualized volumetric parameter for the early detection of clinical benefit of targeted therapies. Ann Oncol. 2016;27(8):1638–1643.
    1. Reynaud T, Bertaut A, Farah W, et al. . Hypofractionated stereotactic radiotherapy as a salvage therapy for recurrent high-grade gliomas: single-center experience. Technol Cancer Res Treat. 2018;17(January – December):1–11.
    1. Randles A, Wirsching H, Dean JA, et al. . Computational modelling of perivascular-niche dynamics for the optimization of treatment schedules for glioblastoma. Nat Biomed Eng. 2021;5(April):346–359.
    1. Fogh SE, Andrews DW, Glass J, et al. . Hypofractionated stereotactic radiation therapy: an effective therapy for recurrent high-grade gliomas. J Clin Oncol. 2010;28(18):3048–3053.
    1. Tsien CI, Pugh SL, Dicker AP, et al. . NRG oncology/RTOG1205: a randomized phase II trial of concurrent bevacizumab and reirradiation versus bevacizumab alone as treatment for recurrent glioblastoma. J Clin Oncol. 2022.
    1. Wong CH, Siah KW, Lo AW. Estimation of clinical trial success rates and related parameters. Biostatistics. 2019;20(2):273–286.
    1. Altrock PM, Liu LL, Michor F. The mathematics of cancer: integrating quantitative models. Nat Rev Cancer. 2015;15(12):730–745.
    1. Combs SE, Thilmann C, Edler L, Debus J, Schulz-Ertner D. Efficacy of fractionated stereotactic reirradiation in recurrent gliomas: long-term results in 172 patients treated in a single institution. J Clin Oncol. 2005;23(34):8863–8869.
    1. Arvold ND, Shi DD, Aizer AA, et al. . Salvage re-irradiation for recurrent high-grade glioma and comparison to bevacizumab alone. J Neurooncol. 2017;135(3):581–591.
    1. Shapiro LQ, Beal K, Goenka A, et al. . Patterns of failure after concurrent bevacizumab and hypofractionated stereotactic radiation therapy for recurrent high-grade glioma. Int J Radiat Oncol Biol Phys. 2013;85(3):636–642.
    1. Vogelbaum MA, Krivosheya D, Borghei-Razavi H, et al. . Phase 0 and window of opportunity clinical trial design in neuro-oncology: a RANO review. Neuro Oncol. 2020;22(11):1568–1579.

Source: PubMed

3
Abonneren