Tumor Drug Concentration and Phosphoproteomic Profiles After Two Weeks of Treatment With Sunitinib in Patients with Newly Diagnosed Glioblastoma

Myra E van Linde, Mariette Labots, Cyrillo G Brahm, Koos E Hovinga, Philip C De Witt Hamer, Richard J Honeywell, Richard de Goeij-de Haas, Alex A Henneman, Jaco C Knol, Godefridus J Peters, Henk Dekker, Sander R Piersma, Thang V Pham, William P Vandertop, Connie R Jiménez, Henk M W Verheul, Myra E van Linde, Mariette Labots, Cyrillo G Brahm, Koos E Hovinga, Philip C De Witt Hamer, Richard J Honeywell, Richard de Goeij-de Haas, Alex A Henneman, Jaco C Knol, Godefridus J Peters, Henk Dekker, Sander R Piersma, Thang V Pham, William P Vandertop, Connie R Jiménez, Henk M W Verheul

Abstract

Purpose: Tyrosine kinase inhibitors (TKI) have poor efficacy in patients with glioblastoma (GBM). Here, we studied whether this is predominantly due to restricted blood-brain barrier penetration or more to biological characteristics of GBM.

Patients and methods: Tumor drug concentrations of the TKI sunitinib after 2 weeks of preoperative treatment was determined in 5 patients with GBM and compared with its in vitro inhibitory concentration (IC50) in GBM cell lines. In addition, phosphotyrosine (pTyr)-directed mass spectrometry (MS)-based proteomics was performed to evaluate sunitinib-treated versus control GBM tumors.

Results: The median tumor sunitinib concentration of 1.9 μmol/L (range 1.0-3.4) was 10-fold higher than in concurrent plasma, but three times lower than sunitinib IC50s in GBM cell lines (median 5.4 μmol/L, 3.0-8.5; P = 0.01). pTyr-phosphoproteomic profiles of tumor samples from 4 sunitinib-treated versus 7 control patients revealed 108 significantly up- and 23 downregulated (P < 0.05) phosphopeptides for sunitinib treatment, resulting in an EGFR-centered signaling network. Outlier analysis of kinase activities as a potential strategy to identify drug targets in individual tumors identified nine kinases, including MAPK10 and INSR/IGF1R.

Conclusions: Achieved tumor sunitinib concentrations in patients with GBM are higher than in plasma, but lower than reported for other tumor types and insufficient to significantly inhibit tumor cell growth in vitro. Therefore, alternative TKI dosing to increase intratumoral sunitinib concentrations might improve clinical benefit for patients with GBM. In parallel, a complex profile of kinase activity in GBM was found, supporting the potential of (phospho)proteomic analysis for the identification of targets for (combination) treatment.

Trial registration: ClinicalTrials.gov NCT02239952.

©2022 The Authors; Published by the American Association for Cancer Research.

Figures

Figure 1.
Figure 1.
Sunitinib effect in vitro. Proliferation (MTT) assay of GBM and colorectal cancer cell lines incubated with increasing sunitinib concentrations, showing the percentage of proliferation compared with untreated controls (left) and calculated IC50 in micromoles per liter (right). Sunitinib inhibited proliferation of GBM tumor cells in vitro at concentrations 2.8 times higher than achieved intratumorally in patients (Table 1). GBM, glioblastoma; CRC, colorectal cancer.
Figure 2.
Figure 2.
Supervised clustering analysis of the tyrosine phosphoproteome. Supervised hierarchical clustering of differential phosphopeptides (P < 0.05) identified by tyrosine-phosphoproteomics shows separation of sunitinib-treated and control tumor tissues from 11 patients with glioblastoma. The heatmap shows the relative phosphopeptide intensities (z-score) in these samples based on log10-transformed values (orange, high abundance; blue, low abundance). SUN, sunitinib-treated; CON, control.
Figure 3.
Figure 3.
Protein interaction network. Protein interaction network of differential phosphopeptides (P < 0.05) between sunitinib-treated and control samples. Regulated phosphopeptides are mapped to proteins and visualized as protein interaction network. Green, Down- and Red, Upregulated in sunitinib-treated patients. Colored subdivisions indicate identification of multiple upregulated phosphopeptides mapping to the same protein. Note EGFR was identified with three upregulated phosphopeptides in the sunitinib-treated group.
Figure 4.
Figure 4.
Ranking of top 20 active kinases in tumors from 11 patients with glioblastoma. Ranked kinase activities in 7 control and 4 sunitinib-treated tumors. For each tumor, bar graphs depict kinase ranking based on combined INKA scores of kinase- and substrate-centric analysis of tyrosine-phoshoproteomics (12). For patient SUN-04, three biological replicates have been analyzed. Top bars of potential hyperactive kinases are highlighted by dark coloring (MAPK10 blue, EGFR red). Note the very high INKA score for MAPK10 in patient CON-08. Light blue and red colored bars indicate lower ranked MAPK10 and EGFR kinase activities, respectively. Green colors highlight kinases with “outlier activity”, i.e., higher activity in one versus the remaining 10 tumors (see Supplementary Fig. S3). SUN, sunitinib-treated; CON, control.

References

    1. De Witt Hamer PC. Small molecule kinase inhibitors in glioblastoma: a systematic review of clinical studies. Neuro Oncol 2010;12:304–16.
    1. Neyns B, Sadones J, Chaskis C, Dujardin M, Everaert H, Lv S, et al. . Phase II study of sunitinib malate in patients with recurrent high-grade glioma. J Neurooncol 2011;103:491–501.
    1. Lee EQ, Kaley TJ, Duda DG, Schiff D, Lassman AB, Wong ET, et al. . A multicenter, phase II, randomized, noncomparative clinical trial of radiation and temozolomide with or without vandetanib in newly diagnosed glioblastoma patients. Clin Cancer Res 2015;21:3610–8.
    1. Thomas AA, Brennan CW, DeAngelis LM, Omuro AM. Emerging therapies for glioblastoma. JAMA Neurol 2014;71:1437–44.
    1. Nakada M, Kita D, Watanabe T, Hayashi Y, Hamada J. Mechanism of chemoresistance against tyrosine kinase inhibitors in malignant glioma. Brain Tumor Pathol 2014;31:198–207.
    1. Pearson JRD, Regad T. Targeting cellular pathways in glioblastoma multiforme. Signal Transduct Target Ther 2017;2:17040.
    1. Fokas E, Steinbach JP, Rodel C. Biology of brain metastases and novel targeted therapies: time to translate the research. Biochim Biophys Acta 2013;1835:61–75.
    1. Klaeger S, Heinzlmeir S, Wilhelm M, Polzer H, Vick B, Koenig PA, et al. . The target landscape of clinical kinase drugs. Science 2017;358:eaan4368.
    1. Rovithi M, Verheul HMW. Pulsatile high-dose treatment with antiangiogenic tyrosine kinase inhibitors improves clinical antitumor activity. Angiogenesis 2017;20:287–9.
    1. Cutillas PR. Role of phosphoproteomics in the development of personalized cancer therapies. Proteomics Clin Appl 2015;9:383–95.
    1. Olsen JV, Vermeulen M, Santamaria A, Kumar C, Miller ML, Jensen LJ, et al. . Quantitative phosphoproteomics reveals widespread full phosphorylation site occupancy during mitosis. Sci Signal 2010;3:ra3.
    1. Beekhof R, van Alphen C, Henneman AA, Knol JC, Pham TV, Rolfs F, et al. . INKA, an integrative data analysis pipeline for phosphoproteomic inference of active kinases. Mol Syst Biol 2019;15:e8250.
    1. Jimenez CR, Verheul HM. Mass spectrometry-based proteomics: from cancer biology to protein biomarkers, drug targets, and clinical applications. Am Soc Clin Oncol Educ Book 2014:e504–10.
    1. Rikova K, Guo A, Zeng Q, Possemato A, Yu J, Haack H, et al. . Global survey of phosphotyrosine signaling identifies oncogenic kinases in lung cancer. Cell 2007;131:1190–203.
    1. Petty WJ, Dragnev KH, Memoli VA, Ma Y, Desai NB, Biddle A, et al. . Epidermal growth factor receptor tyrosine kinase inhibition represses cyclin D1 in aerodigestive tract cancers. Clin Cancer Res 2004;10:7547–54.
    1. Labots M, Pham TV, Honeywell RJ, Knol JC, Beekhof R, de Goeij-de Haas R, et al. . Kinase inhibitor treatment of patients with advanced cancer results in high tumor drug concentrations and in specific alterations of the tumor phosphoproteome. Cancers 2020;12:330.
    1. Labots M, van der Mijn JC, Beekhof R, Piersma SR, de Goeij-de Haas RR, Pham TV, et al. . Phosphotyrosine-based-phosphoproteomics scaled-down to biopsy level for analysis of individual tumor biology and treatment selection. J Proteomics 2017;162:99–107.
    1. Gotink KJ, Rovithi M, de Haas RR, Honeywell RJ, Dekker H, Poel D, et al. . Cross-resistance to clinically used tyrosine kinase inhibitors sunitinib, sorafenib and pazopanib. Cell Oncol 2015;38:119–29.
    1. Gotink KJ, Broxterman HJ, Labots M, de Haas RR, Dekker H, Honeywell RJ, et al. . Lysosomal sequestration of sunitinib: a novel mechanism of drug resistance. Clin Cancer Res 2011;17:7337–46.
    1. Piersma SR, Knol JC, de Reus I, Labots M, Sampadi BK, Pham TV, et al. . Feasibility of label-free phosphoproteomics and application to base-line signaling of colorectal cancer cell lines. J Proteomics 2015;127:247–58.
    1. van der Mijn JC, Labots M, Piersma SR, Pham TV, Knol JC, Broxterman HJ, et al. . Evaluation of different phospho-tyrosine antibodies for label-free phosphoproteomics. J Proteomics 2015;127:259–63.
    1. Cox J, Mann M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat Biotechnol 2008;26:1367–72.
    1. Krug K, Mertins P, Zhang B, Hornbeck P, Raju R, Ahmad R, et al. . A curated resource for phosphosite-specific signature analysis. Mol Cell Proteomics 2019;18:576–93.
    1. Vizcaino JA, Deutsch EW, Wang R, Csordas A, Reisinger F, Rios D, et al. . ProteomeXchange provides globally coordinated proteomics data submission and dissemination. Nat Biotechnol 2014;32:223–6.
    1. Gotink KJ, Broxterman HJ, Honeywell RJ, Dekker H, de Haas RR, Miles KM, et al. . Acquired tumor cell resistance to sunitinib causes resistance in a HT-29 human colon cancer xenograft mouse model without affecting sunitinib biodistribution or the tumor microvasculature. Oncoscience 2014;1:844–53.
    1. Honeywell RJ, Kathmann I, Giovannetti E, Tibaldi C, Smit EF, Rovithi MN, et al. . Epithelial transfer of the tyrosine kinase inhibitors erlotinib, gefitinib, afatinib, crizotinib, sorafenib, sunitinib, and dasatinib: implications for clinical resistance. Cancers 2020;12:3322.
    1. Heffron TP. Challenges of developing small-molecule kinase inhibitors for brain tumors and the need for emphasis on free drug levels. Neuro Oncol 2018;20:307–12.
    1. van Tellingen O, Yetkin-Arik B, de Gooijer MC, Wesseling P, Wurdinger T, de Vries HE. Overcoming the blood-brain tumor barrier for effective glioblastoma treatment. Drug Resist Updat 2015;19:1–12.
    1. Deng J, Shao J, Markowitz JS, An G. ABC transporters in multi-drug resistance and ADME-Tox of small molecule tyrosine kinase inhibitors. Pharm Res 2014;31:2237–55.
    1. Parrish KE, Cen L, Murray J, Calligaris D, Kizilbash S, Mittapalli RK, et al. . Efficacy of PARP inhibitor rucaparib in orthotopic glioblastoma xenografts is limited by ineffective drug penetration into the central nervous system. Mol Cancer Ther 2015;14:2735–43.
    1. Pokorny JL, Calligaris D, Gupta SK, Iyekegbe DO Jr, Mueller D, Bakken KK, et al. . The efficacy of the Wee1 inhibitor MK-1775 combined with temozolomide is limited by heterogeneous distribution across the blood-brain barrier in glioblastoma. Clin Cancer Res 2015;21:1916–24.
    1. Laramy JK, Kim M, Gupta SK, Parrish KE, Zhang S, Bakken KK, et al. . Heterogeneous binding and central nervous system distribution of the multitargeted kinase inhibitor ponatinib restrict orthotopic efficacy in a patient-derived xenograft model of glioblastoma. J Pharmacol Exp Ther 2017;363:136–47.
    1. Plotkin S, Supko JG, Curry WT. Concentrations of Sunitinib after oral administration in patients with high grade glioma [abstract]. Neuro-oncol 2011;13:iii41–68.
    1. Abbruzzese C, Matteoni S, Signore M, Cardone L, Nath K, Glickson JD, et al. . Drug repurposing for the treatment of glioblastoma multiforme. J Exp Clin Cancer Res 2017;36:169.
    1. Lankheet NAG, Desar IME, Mulder SF, Burger DM, Kweekel DM, van Herpen CML, et al. . Optimizing the dose in cancer patients treated with imatinib, sunitinib and pazopanib. Br J Clin Pharmacol 2017;83:2195–204.
    1. Rovithi M, Gerritse SL, Honeywell RJ, Ten Tije AJ, Ruijter R, Peters GJ, et al. . Phase I dose-escalation study of once weekly or once every two weeks administration of high-dose sunitinib in patients with refractory solid tumors. J Clin Oncol 2019;37:411–8.
    1. Zhu Y, Du Y, Liu H, Ma T, Shen Y, Pan Y. Study of efficacy and safety of pulsatile administration of high-dose gefitinib or erlotinib for advanced non-small cell lung cancer patients with secondary drug resistance: a single center, single arm, phase II clinical trial. Thorac Cancer 2016;7:663–9.
    1. Morikawa A, de Stanchina E, Pentsova E, Kemeny MM, Li BT, Tang K, et al. . Phase I study of intermittent high-dose lapatinib alternating with capecitabine for HER2-positive breast cancer patients with central nervous system metastases. Clin Cancer Res 2019;25:3784–92.
    1. Kim DH, Choi YJ, Sung KJ, Yoo SA, Sung YH, Kim JK, et al. . Efficacy of nano-particulated, water-soluble erlotinib against intracranial metastases of EGFR-mutant lung cancer. Mol Oncol 2018;12:2182–90.
    1. Sharma S, Petsalaki E. Large-scale datasets uncovering cell signalling networks in cancer: context matters. Curr Opin Genet Dev 2019;54:118–24.
    1. Sever R, Brugge JS. Signal transduction in cancer. Cold Spring Harb Perspect Med 2015;5:a006098.
    1. Johnson H, White FM. Quantitative analysis of signaling networks across differentially embedded tumors highlights interpatient heterogeneity in human glioblastoma. J Proteome Res 2014;13:4581–93.
    1. Randall EC, Emdal KB, Laramy JK, Kim M, Roos A, Calligaris D, et al. . Integrated mapping of pharmacokinetics and pharmacodynamics in a patient-derived xenograft model of glioblastoma. Nat Commun 2018;9:4904.
    1. Wei W, Shin YS, Xue M, Matsutani T, Masui K, Yang H, et al. . Single-cell phosphoproteomics resolves adaptive signaling dynamics and informs targeted combination therapy in glioblastoma. Cancer Cell 2016;29:563–73.
    1. Davis RJ. Signal transduction by the JNK group of MAP kinases. Cell 2000;103:239–52.
    1. Nakano R, Nakayama T, Sugiya H. Biological properties of JNK3 and its function in neurons, astrocytes, pancreatic beta-cells and cardiovascular cells. Cells 2020;9:1802.
    1. Antonyak MA, Kenyon LC, Godwin AK, James DC, Emlet DR, Okamoto I, et al. . Elevated JNK activation contributes to the pathogenesis of human brain tumors. Oncogene 2002;21:5038–46.
    1. Cui J, Han SY, Wang C, Su W, Harshyne L, Holgado-Madruga M, et al. . c-Jun NH(2)-terminal kinase 2alpha2 promotes the tumorigenicity of human glioblastoma cells. Cancer Res 2006;66:10024–31.
    1. Koch P, Gehringer M, Laufer SA. Inhibitors of c-Jun N-terminal kinases: an update. J Med Chem 2015;58:72–95.
    1. Gong Y, Ma Y, Sinyuk M, Loganathan S, Thompson RC, Sarkaria JN, et al. . Insulin-mediated signaling promotes proliferation and survival of glioblastoma through Akt activation. Neuro Oncol 2016;18:48–57.

Source: PubMed

3
購読する