Sequential bortezomib and temozolomide treatment promotes immunological responses in glioblastoma patients with positive clinical outcomes: A phase 1B study

Mohummad A Rahman, Jorunn Brekke, Victoria Arnesen, Marianne H Hannisdal, Andrea G Navarro, Andreas Waha, Lars Herfindal, Cecilie B Rygh, Eirik Bratland, Petter Brandal, Judit Haasz, Leif Oltedal, Hrvoje Miletic, Arvid Lundervold, Stein A Lie, Dorota Goplen, Martha Chekenya, Mohummad A Rahman, Jorunn Brekke, Victoria Arnesen, Marianne H Hannisdal, Andrea G Navarro, Andreas Waha, Lars Herfindal, Cecilie B Rygh, Eirik Bratland, Petter Brandal, Judit Haasz, Leif Oltedal, Hrvoje Miletic, Arvid Lundervold, Stein A Lie, Dorota Goplen, Martha Chekenya

Abstract

Background: Glioblastoma (GBM) is an aggressive malignant brain tumor where median survival is approximately 15 months after best available multimodal treatment. Recurrence is inevitable, largely due to O6 methylguanine DNA methyltransferase (MGMT) that renders the tumors resistant to temozolomide (TMZ). We hypothesized that pretreatment with bortezomib (BTZ) 48 hours prior to TMZ to deplete MGMT levels would be safe and tolerated by patients with recurrent GBM harboring unmethylated MGMT promoter. The secondary objective was to investigate whether 26S proteasome blockade may enhance differentiation of cytotoxic immune subsets to impact treatment responses measured by radiological criteria and clinical outcomes.

Methods: Ten patients received intravenous BTZ 1.3 mg/m2 on days 1, 4, and 7 during each 4th weekly TMZ-chemotherapy starting on day 3 and escalated from 150 mg/m2 per oral 5 days/wk via 175 to 200 mg/m2 in cycles 1, 2, and 3, respectively. Adverse events and quality of life were evaluated by CTCAE and EQ-5D-5L questionnaire, and immunological biomarkers evaluated by flow cytometry and Luminex enzyme-linked immunosorbent assay.

Results: Sequential BTZ + TMZ therapy was safe and well tolerated. Pain and performance of daily activities had greatest impact on patients' self-reported quality of life and were inversely correlated with Karnofsky performance status. Patients segregated a priori into three groups, where group 1 displayed stable clinical symptoms and/or slower magnetic resonance imaging radiological progression, expanded CD4+ effector T-cells that attenuated cytotoxic T-lymphocyte associated protein-4 and PD-1 expression and secreted interferon γ and tumor necrosis factor α in situ and ex vivo upon stimulation with PMA/ionomycin. In contrast, rapidly progressing group 2 patients exhibited tolerised T-cell phenotypes characterized by fourfold to sixfold higher interleukin 4 (IL-4) and IL-10 Th-2 cytokines after BTZ + TMZ treatment, where group 3 patients exhibited intermediate clinical/radiological responses.

Conclusion: Sequential BTZ + TMZ treatment is safe and promotes Th1-driven immunological responses in selected patients with improved clinical outcomes (Clinicaltrial.gov (NCT03643549)).

Keywords: MGMT; Th1/Th2 cytokine ratios; bortezomib; immune checkpoint; recurrent GBM; temozolomide.

Conflict of interest statement

The authors declare that there are no conflict of interests.

© 2020 The Authors. Immunity, Inflammation and Disease published by John Wiley & Sons Ltd.

Figures

Figure 1
Figure 1
Schematic of trial schedule. A, Timeline showing BORTEM‐17 treatment regimen. Bortezomib administered intravenous at days 1, 4, and 7 (48 hours pretreatment to deplete MGMT protein) before target TMZ 200 mg/m2 dose for 5 days (from and including days 3‐7), repeated in six cycles. In n = 3 patients per each dose 150 vs 175 vs 200 mg/m2 TMZ in dose pathfinding, safety evaluation. Clinical chemistry for renal, hepatic, and bone marrow monitoring for toxicity based on CTCAE v. 4.03. MRI tumor monitoring radiological response assessment based on RANO criteria. Rationale for sequential treatment schedule based on preclinical data. B, sequential administration in BORTEM‐17 clinical trial vs (C) previous studies where BTZ on days 1, 4, 8, and 11 was given concomitantly with TMZ from day 1 to 5, when MGMT levels were high. Dashed boxes mark days when TMZ doses might be more effective, (B) all five doses vs (C) three doses every month. BTZ, bortezomib; CBC Diff, complete blood count with differential test; CMP, comprehensive metabolic panel; KPS, Karnofsky performance score; LFT, liver function test; MGMT, O6‐methyl guanine DNA methyltransferase; MRI, magnetic resonance imaging; NANO, neurologic assessment in neuro‐oncology; QoL, quality of life; TMZ, temozolomide
Figure 2
Figure 2
Clinical, radiological and quality of life response. A, Pharmacokinetic analysis of bortezomib concentration (ng/mL) in plasma of patients following five timepoints on day 1 and 7 of treatment. B, Thrombocyte counts during the first three treatment cycles. Data represent the mean ± SEM from n = 10 patients. C, Density scores for perception of overall health on EQ‐5D‐5L questionnaire. D, Correlation of self‐reported overall health and levels of pain on EQ‐5D‐5L. E, Swimmer plot of individual patients depicting treatment start/stop times of all trial patients, aligned according to their first surgery. Bars represent survival in months from first surgery, first and second MRI progression. Rightward arrow indicates that the patient was still alive at the time of final data collection
Figure 3
Figure 3
Tumour growth and population doubling time. A, Coregistered 3D T1 weighted gadolinium contrast enhanced serial MR image of patient‐03 (IDHwt; MGMT UM, 54 years male) treated for 6 months, time indicated in days and tumour volume in µL. B, Mean of 3D measured tumour volumes in mm3 from T1‐weighted MR images with contrast of all patients, and (C) tumor population doubling time (in days) for group 1, 2, and 3 patients
Figure 4
Figure 4
Immune mechanisms and augmented differentiation of CD4+ and CD8+ T cell subsets in patients with positive clinical responses. A, Cytokine concentrations in pg/mL in patients' plasma, (top panels) IL‐4 and IL‐10, and (bottom panels) the ratio between the average concentrations of IL‐5 and IL‐4 against IFNγ during treatment. B, Representative dot plots showing (from left to right) patient NK and T cell lymphocytes in CD56 vs CD3 gates, CD8+ vs CD4+ T cell subsets and CD3+CD4+ T cells expressing CTLA‐4. C, Representative dot plots showing CD3+CD4+ T cell subsets expressing CTLA‐4 and (D) Representative dot plots showing CD3+CD8+ T cell subsets expressing PD‐1. E, Mean ± SEM % of CD3+CD4+ T cells expression of PD‐1 and CTLA‐4 in group 1, 2, and 3 patients before, during and after treatment with BTZ and TMZ. F, Mean ± SEM % of CD3+CD8+ expressing PD‐1 and CTLA‐4 before and after treatment with BTZ and TMZ. G, Dot plots showing CCR7 vs CD45RO within CD3+CD4+ T cell subsets in group 1, 2, and 3 patients, before and during treatment with BTZ and TMZ. H, Mean ± SEM % expression of naïve (N), effector (E), central memory (CM) and effector memory (EM) CD4+ T‐cells, and (I) CD4+ T‐effector cells before, during and after treatment with BTZ and TMZ in group 1, 2, and 3 patients. *P < .05, **P < .01, ***P < .001, ****P < .0001, n = 10 patients. BTZ, bortezomib; IFNγ, interferon γ; IL, interleukin; NK, natural killer; TMZ, temozolomide
Figure 5
Figure 5
PBMCs from patients with positive clinical responses diminish CTLA‐4+ CD4+Th cells, attenuate IL‐10 secretion and have moderate intracellular IFNγ after bortezomib + temozolomide treatment. A, Representative dot plots showing CD4 vs CTLA‐4 in patient PBMCs unstimulated and stimulated, with PMA/ionomycin before or after exposure to BTZ or BTZ + TMZ treatment in situ. B, Mean ± SEM % of CD4+ CTLA‐4+ T cells at baseline or after treatment/stimulation conditions in group 1, 2, and 3 patients. C, Mean ± SEM % of CD8+ CTLA‐4+ at baseline or after treatment/stimulation conditions in group 1, 2, and 3 patients. D Representative dot plots showing intracellular IFNγ in CD8+T cells in patients' PBMCs unstimulated and stimulated with PMA/ionomycin in group 1 vs 2 patients. E, Mean ± SEM % of CD8+ IFNγ+ T cells at baseline or after treatment/stimulation conditions in group 1, 2, and 3 patients. Cytokines present in supernatants from patients' PBMCs after stimulation with PMA/ionomycin represented as mean ± SEM of Th2/Th1 ratio between (F) IL‐10 and IFNγ, (G) IL‐10 and TNF‐α, and (H) IL‐4 and TNF‐α in group 1, 2, and 3 patients at baseline or during treatment recovery. I, Mean ± SEM % expression of markers in patient‐02 tumour or PBMCs unstimulated and stimulated with PMA/ionomycin or P02‐tumour before or after exposure to bortezomib + temozolomide in situ. Cytokine concentrations in pg/mL from supernatants from patient‐02 tumour or PBMCs unstimulated or stimulated ex vivo with PMA + ionomycin or patient‐02 tumour or before and after exposure to bortezomib + temozolomide in situ of (J) IL‐2 and (K) TNF‐α. *P < .05; **P < .01; ***P < .001: ****P < .0001, n = 10 patients. BTZ, bortezomib; IFNγ, interferon γ; IL, interleukin; TMZ, temozolomide; TNF‐α, tumor necrosis factor α

References

    1. Sanai N, Polley MY, McDermott MW, Parsa AT, Berger MS. An extent of resection threshold for newly diagnosed glioblastomas. J Neurosurg. 2011;115:3‐8. 10.3171/2011.2.JNS10998
    1. Stummer W, Pichlmeier U, Meinel T, et al. Fluorescence‐guided surgery with 5‐aminolevulinic acid for resection of malignant glioma: a randomised controlled multicentre phase III trial. Lancet Oncol. 2006;7:392‐401. 10.1016/S1470-2045(06)70665-9
    1. Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352:987‐996. 10.1056/NEJMoa043330
    1. Chinot OL, Wick W, Mason W, et al. Bevacizumab plus radiotherapy‐temozolomide for newly diagnosed glioblastoma. N Engl J Med. 2014;370:709‐722. 10.1056/NEJMoa1308345
    1. Gilbert MR, Dignam JJ, Armstrong TS, et al. A randomized trial of bevacizumab for newly diagnosed glioblastoma. N Engl J Med. 2014;370:699‐708. 10.1056/NEJMoa1308573
    1. Hegi ME, Diserens AC, Gorlia T, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med. 2005;352:997‐1003. 10.1056/NEJMoa043331
    1. Popescu RA, Schäfer R, Califano R, et al. The current and future role of the medical oncologist in the professional care for cancer patients: a position paper by the European Society for Medical Oncology (ESMO). Ann Oncol. 2014;25:9‐15. 10.1093/annonc/mdt522
    1. Louis DN, Perry A, Reifenberger G, et al. The 2016 world health organization classification of tumors of the central nervous system: a summary. Acta Neuropathol. 2016;131:803‐820. 10.1007/s00401-016-1545-1
    1. Ohgaki H, Kleihues P. The definition of primary and secondary glioblastoma. Clin Cancer Res. 2013;19:764‐772. 10.1158/1078-0432.CCR-12-3002
    1. Wick W, Hartmann C, Engel C, et al. NOA‐04 randomized phase III trial of sequential radiochemotherapy of anaplastic glioma with procarbazine, lomustine, and vincristine or temozolomide. J Clin Oncol. 2009;27:5874‐5880. 10.1200/JCO.2009.23.6497
    1. Quinn JA, Desjardins A, Weingart J, et al. Phase I trial of temozolomide plus O6‐benzylguanine for patients with recurrent or progressive malignant glioma. J Clin Oncol. 2005;23:7178‐7187. 10.1200/JCO.2005.06.502
    1. Quinn JA, Pluda J, Dolan ME, et al. Phase II trial of carmustine plus O(6)‐benzylguanine for patients with nitrosourea‐resistant recurrent or progressive malignant glioma. J Clin Oncol. 2002;20:2277‐2283.
    1. Viel T, Monfared P, Schelhaas S, et al. Optimizing glioblastoma temozolomide chemotherapy employing lentiviral‐based anti‐MGMT shRNA technology. Mol Ther. 2013;21:570‐579. 10.1038/mt.2012.278
    1. Weingart J, Grossman SA, Carson KA, et al. Phase I trial of polifeprosan 20 with carmustine implant plus continuous infusion of intravenous O6‐benzylguanine in adults with recurrent malignant glioma: new approaches to brain tumor therapy CNS consortium trial. J Clin Oncol. 2007;25:399‐404. 10.1200/JCO.2006.06.6290
    1. Lavon I, Fuchs D, Zrihan D, et al. Novel mechanism whereby nuclear factor kappaB mediates DNA damage repair through regulation of O(6)‐methylguanine‐DNA‐methyltransferase. Cancer Res. 2007;67:8952‐8959. 10.1158/0008-5472.CAN-06-3820
    1. Bocangel D, Sengupta S, Mitra S, Bhakat KK. p53‐Mediated down‐regulation of the human DNA repair gene O6‐methylguanine‐DNA methyltransferase (MGMT) via interaction with Sp1 transcription factor. Anticancer Res. 2009;29:3741‐3750.
    1. Harris LC, Remack JS, Houghton PJ, Brent TP. Wild‐type p53 suppresses transcription of the human O6‐methylguanine‐DNA methyltransferase gene. Cancer Res. 1996;56:2029‐2032.
    1. Persano L, Pistollato F, Rampazzo E, et al. BMP2 sensitizes glioblastoma stem‐like cells to Temozolomide by affecting HIF‐1alpha stability and MGMT expression. Cell Death Dis. 2012;3:e412 10.1038/cddis.2012.153
    1. Bhakat KK, Mitra S. Regulation of the human O(6)‐methylguanine‐DNA methyltransferase gene by transcriptional coactivators cAMP response element‐binding protein‐binding protein and p300. J Biol Chem. 2000;275:34197‐34204. 10.1074/jbc.M005447200
    1. Chen FY, Harris LC, Remack JS, Brent TP. Cytoplasmic sequestration of an O6‐methylguanine‐DNA methyltransferase enhancer binding protein in DNA repair‐deficient human cells. Proc Natl Acad Sci USA. 1997;94:4348‐4353. 10.1073/pnas.94.9.4348
    1. Kohsaka S, Wang L, Yachi K, et al. STAT3 inhibition overcomes temozolomide resistance in glioblastoma by downregulating MGMT expression. Mol Cancer Ther. 2012;11:1289‐1299. 10.1158/1535-7163.MCT-11-0801
    1. Rahman MA, Gras Navarro A, Brekke J, et al. Bortezomib administered prior to temozolomide depletes MGMT, chemosensitizes glioblastoma with unmethylated MGMT promoter and prolongs animal survival. Br J Cancer. 2019;10:545‐555. 10.1038/s41416-019-0551-1
    1. Sun SC. The non‐canonical NF‐kappaB pathway in immunity and inflammation. Nat Rev Immunol. 2017;17:545‐558. 10.1038/nri.2017.52
    1. Gras Navarro A, Espedal H, Joseph JV, et al. Pretreatment of glioblastoma with bortezomib potentiates natural killer cell cytotoxicity through TRAIL/DR5 mediated apoptosis and prolongs animal survival Cancers, 2019:11 10.3390/cancers11070996
    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;28:1963‐1972. 10.1200/JCO.2009.26.3541
    1. Nayak L, DeAngelis LM, Brandes AA, et al. The neurologic assessment in neuro‐oncology (NANO) scale: a tool to assess neurologic function for integration into the response assessment in neuro‐oncology (RANO) criteria. Neuro Oncol. 2017;19:625‐635. 10.1093/neuonc/nox029
    1. Brooks R. EuroQol: the current state of play. Health Policy. 1996;37:53‐72.
    1. Dominguez–Valentin M, Gras Navarro A, Rahman AM, et al. Identification of a natural killer cell receptor allele that prolongs survival of cytomegalovirus‐positive glioblastoma patients. Cancer Res. 2016;76:5326‐5336. 10.1158/0008-5472.CAN-16-1162
    1. Reece DE, Sullivan D, Lonial S, et al. Pharmacokinetic and pharmacodynamic study of two doses of bortezomib in patients with relapsed multiple myeloma. Cancer Chemother Pharmacol. 2011;67:57‐67. 10.1007/s00280-010-1283-3
    1. Hanley MJ, Mould DR, Taylor TJ, et al. Population pharmacokinetic analysis of bortezomib in pediatric leukemia patients: model‐based support for body surface area‐based dosing over the 2‐ to 16‐year age range. J Clin Pharmacol. 2017;57:1183‐1193. 10.1002/jcph.906
    1. Chang CL, Hsu YT, Wu CC, et al. Immune mechanism of the antitumor effects generated by bortezomib. J Immunol. 2012;189:3209‐3220. 10.4049/jimmunol.1103826
    1. Schumacher LY, Vo DD, Garban HJ, et al. Immunosensitization of tumor cells to dendritic cell‐activated immune responses with the proteasome inhibitor bortezomib (PS‐341, Velcade). J Immunol. 2006;176:4757‐4765. 10.4049/jimmunol.176.8.4757
    1. Kong XT, Nguyen NT, Choi YJ, et al. Phase 2 study of bortezomib combined with temozolomide and regional radiation therapy for upfront treatment of patients with newly diagnosed glioblastoma multiforme: safety and efficacy assessment. Int J Radiat Oncol Biol Phys. 2018;100:1195‐1203. 10.1016/j.ijrobp.2018.01.001
    1. Kubicek GJ, Werner‐Wasik M, Machtay M, et al. Phase I trial using proteasome inhibitor bortezomib and concurrent temozolomide and radiotherapy for central nervous system malignancies. Int J Radiat Oncol Biol Phys. 2009;74:433‐439. 10.1016/j.ijrobp.2008.08.050
    1. McCracken DJ, Celano EC, Voloschin AD, Read WL, Olson JJ. Phase I trial of dose‐escalating metronomic temozolomide plus bevacizumab and bortezomib for patients with recurrent glioblastoma. J Neurooncol. 2016;130:193‐201. 10.1007/s11060-016-2234-6
    1. Phuphanich S, Supko JG, Carson KA, et al. Phase 1 clinical trial of bortezomib in adults with recurrent malignant glioma. J Neurooncol. 2010;100:95‐103. 10.1007/s11060-010-0143-7
    1. Portnow J, Frankel P, Koehler S, et al. A phase I study of bortezomib and temozolomide in patients with advanced solid tumors. Cancer Chemother Pharmacol. 2012;69:505‐514. 10.1007/s00280-011-1721-x
    1. Odia Y, Kreisl TN, Aregawi D, Innis EK, Fine HA. A phase II trial of tamoxifen and bortezomib in patients with recurrent malignant gliomas. J Neurooncol. 2015;125:191‐195. 10.1007/s11060-015-1894-y
    1. Raizer JJ, Chandler JP, Ferrarese R, et al. A phase II trial evaluating the effects and intra‐tumoral penetration of bortezomib in patients with recurrent malignant gliomas. J Neurooncol. 2016;129:139‐146. 10.1007/s11060-016-2156-3
    1. Nord E. EuroQol: health‐related quality of life measurement. Valuations of health states by the general public in Norway. Health Policy. 1991;18:25‐36. 10.1016/0168-8510(91)90141-j
    1. Jakola AS, Unsgard G, Solheim O. Quality of life in patients with intracranial gliomas: the impact of modern image‐guided surgery. J Neurosurg. 2011;114:1622‐1630. 10.3171/2011.1.JNS101657
    1. Sanson M, Marie Y, Paris S, et al. Isocitrate dehydrogenase 1 codon 132 mutation is an important prognostic biomarker in gliomas. J Clin Oncol. 2009;27:4150‐4154. 10.1200/JCO.2009.21.9832
    1. Neftel C, Laffy J, Filbin MG, et al. An integrative model of cellular states, plasticity, and genetics for glioblastoma. Cell. 2019;178:835‐849 e821. 10.1016/j.cell.2019.06.024
    1. Phillips HS, Kharbanda S, Chen R, et al. Molecular subclasses of high‐grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell. 2006;9:157‐173. 10.1016/j.ccr.2006.02.019
    1. Verhaak RGW, Hoadley KA, Purdom E, et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell. 2010;17:98‐NF110. 10.1016/j.ccr.2009.12.020
    1. Wang Q, Hu B, Hu X, et al. Tumor evolution of glioma‐intrinsic gene expression subtypes associates with immunological changes in the microenvironment. Cancer Cell. 2017;32:42‐56 e46. 10.1016/j.ccell.2017.06.003
    1. Filella X, Alcover J, Zarco MA, Beardo P, Molina R, Ballesta AM. Analysis of type T1 and T2 cytokines in patients with prostate cancer. Prostate. 2000;44:271‐274. 10.1002/1097-0045(20000901)44:4<271::aid-pros2>;2-g
    1. Huang M, Wang J, Lee P, et al. Human non‐small cell lung cancer cells express a type 2 cytokine pattern. Cancer Res. 1995;55:3847‐3853.
    1. Petersson M, Charo J, Salazar‐Onfray F, et al. Constitutive IL‐10 production accounts for the high NK sensitivity, low MHC class I expression, and poor transporter associated with antigen processing (TAP)‐1/2 function in the prototype NK target YAC‐1. J Immunol. 1998;161:2099‐2105.
    1. Mocellin S, Panelli MC, Wang E, Nagorsen D, Marincola FM. The dual role of IL‐10. Trends Immunol. 2003;24:36‐43. 10.1016/s1471-4906(02)00009-1
    1. Conrad CT, Ernst NR, Dummer W, Brocker EB, Becker JC. Differential expression of transforming growth factor beta 1 and interleukin 10 in progressing and regressing areas of primary melanoma. J Exp Clin Cancer Res. 1999;18:225‐232.
    1. Lauw FN, Pajkrt D, Hack CE, Kurimoto M, van Deventer SJ, van der Poll T. Proinflammatory effects of IL‐10 during human endotoxemia. J Immunol. 2000;165:2783‐2789. 10.4049/jimmunol.165.5.2783
    1. Mocellin S, Panelli M, Wang E, et al. IL‐10 stimulatory effects on human NK cells explored by gene profile analysis. Genes Immun. 2004;5:621‐630. 10.1038/sj.gene.6364135
    1. Kidd P. Th1/Th2 balance: the hypothesis, its limitations, and implications for health and disease. Altern Med Rev. 2003;8:223‐246.
    1. Buchbinder EI, Desai A. CTLA‐4 and PD‐1 pathways: similarities, differences, and implications of their inhibition. Am J Clin Oncol. 2016;39:98‐106. 10.1097/COC.0000000000000239
    1. Chambers CA, Sullivan TJ, Truong T, Allison JP. Secondary but not primary T cell responses are enhanced in CTLA‐4‐deficient CD8+ T cells. Eur J Immunol. 1998;28:3137‐3143. 10.1002/(SICI)1521-4141(199810)28:10<3137::AID-IMMU3137>;2-X
    1. Sengupta S, Marrinan J, Frishman C, Sampath P. Impact of temozolomide on immune response during malignant glioma chemotherapy. Clin Dev Immunol. 2012;2012:831090‐831097. 10.1155/2012/831090
    1. Kmiecik J, Poli A, Brons NH, et al. Elevated CD3+ and CD8+ tumor‐infiltrating immune cells correlate with prolonged survival in glioblastoma patients despite integrated immunosuppressive mechanisms in the tumor microenvironment and at the systemic level. J Neuroimmunol. 2013;264:71‐83. 10.1016/j.jneuroim.2013.08.013

Source: PubMed

3
Subskrybuj