Intertumoral heterogeneity in patient-specific drug sensitivities in treatment-naïve glioblastoma

Erlend Skaga, Evgeny Kulesskiy, Artem Fayzullin, Cecilie J Sandberg, Swapnil Potdar, Aija Kyttälä, Iver A Langmoen, Aki Laakso, Emília Gaál-Paavola, Markus Perola, Krister Wennerberg, Einar O Vik-Mo, Erlend Skaga, Evgeny Kulesskiy, Artem Fayzullin, Cecilie J Sandberg, Swapnil Potdar, Aija Kyttälä, Iver A Langmoen, Aki Laakso, Emília Gaál-Paavola, Markus Perola, Krister Wennerberg, Einar O Vik-Mo

Abstract

Background: A major barrier to effective treatment of glioblastoma (GBM) is the large intertumoral heterogeneity at the genetic and cellular level. In early phase clinical trials, patient heterogeneity in response to therapy is commonly observed; however, how tumor heterogeneity is reflected in individual drug sensitivities in the treatment-naïve glioblastoma stem cells (GSC) is unclear.

Methods: We cultured 12 patient-derived primary GBMs as tumorspheres and validated tumor stem cell properties by functional assays. Using automated high-throughput screening (HTS), we evaluated sensitivity to 461 anticancer drugs in a collection covering most FDA-approved anticancer drugs and investigational compounds with a broad range of molecular targets. Statistical analyses were performed using one-way ANOVA and Spearman correlation.

Results: Although tumor stem cell properties were confirmed in GSC cultures, their in vitro and in vivo morphology and behavior displayed considerable tumor-to-tumor variability. Drug screening revealed significant differences in the sensitivity to anticancer drugs (p < 0.0001). The patient-specific vulnerabilities to anticancer drugs displayed a heterogeneous pattern. They represented a variety of mechanistic drug classes, including apoptotic modulators, conventional chemotherapies, and inhibitors of histone deacetylases, heat shock proteins, proteasomes and different kinases. However, the individual GSC cultures displayed high biological consistency in drug sensitivity patterns within a class of drugs. An independent laboratory confirmed individual drug responses.

Conclusions: This study demonstrates that patient-derived and treatment-naïve GSC cultures maintain patient-specific traits and display intertumoral heterogeneity in drug sensitivity to anticancer drugs. The heterogeneity in patient-specific drug responses highlights the difficulty in applying targeted treatment strategies at the population level to GBM patients. However, HTS can be applied to uncover patient-specific drug sensitivities for functional precision medicine.

Keywords: Drug sensitivity; Functional precision medicine; Glioblastoma; Glioblastoma stem cells; High-throughput drug screening; Individualized medicine.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Characterization of patient-derived GSCs. Magnetic resonance imaging of four GBMs in the study cohort (a) and the corresponding xenografts (b) demonstrating that GSC cultures established from a heterogeneous GBM population display culture-to-culture heterogeneity in their in vivo formation characteristics. Images in (b) are stained with Hematoxylin & Eosin (h&e) in the upper image and Hoechst 33258 in the lower image. Tumor borders are macro-anatomically delineated. Scale bar 1 mm. (c) All histopathological features of glioblastoma were identified, including pathological angiogenesis (whole arrow), intratumoral hemorrhages (dotted arrow), tumor necrosis (triangle), pseudopalisading (asterisk) and nuclear atypia with aberrant mitoses. All tumors were xenografted to ≥2 mice. (d) Upon differentiation, the cells displayed a more mature morphology and stained positive for nestin and GFAP, however the individual GSC culture displayed intertumoral variability in their differentiation morphology. Scale bar 50 μm. (e) The cultures displayed variability in their capacity for total cell yield following serial passages, and (f) intertumoral heterogeneity in expression of stem cell markers (f). Expression of stem cell markers are data generated from n = 1 experiments in the individual cultures
Fig. 2
Fig. 2
GSC sensitivity to anticancer drugs. (a) Presentation of four drug responses from the DSRT to the FDA-approved protein synthesis inhibitor omacetaxine. The dose-response curves and DSS demonstrate a drug response below the threshold defined as moderate activity (DSS ≥10) and three other responses with increasing efficacy from moderate to very strong. (b) Number of drugs from the DSRT in each GSC culture with a DSS ≥10. (c) Significance table of the distribution of the overall drug sensitivity to the drug collection (n = 461 drugs) in the primary GSC cultures. Using a non-parametric one-way ANOVA of ranks corrected for multiple comparisons, a significant difference was observed in the overall drug sensitivity (p < 0.0001). (d) Correspondence analysis of all drug responses displays a clear separation of the two most sensitive cultures along the first component variance (14.9%), whereas no identified pattern explained the spread of the cultures along the second component variance (11.3%). Each dot in the scatter plot represents individual drugs (rows), while individual tumors are highlighted (columns)
Fig. 3
Fig. 3
Drug sensitivity in primary GSCs across different drug classes and molecular targets. The figure displays drug class, the drug sensitivity in GSC cultures, and average (± SD) Spearman’s coefficient (ρ) from correlation matrices for drug categories that were represented with ≥3 drugs for the specific molecular target (n = 47 drugs in the figure, all drug sensitivity data in Additional file 3). Correlation matrices demonstrated that the sensitivity to a drug within a category was strongly associated with sensitivity to all other drugs within that drug category, demonstrating biological consistency and individual uniqueness in GSC cultures. Highlighted in red and blue are the tumors found with the highest and lowest sensitivity within the specified category, respectively
Fig. 4
Fig. 4
Unsupervised hierarchical clustering of drug sensitivity patterns in primary GBM and relation to subtype and MGMT status. Heat map and unsupervised hierarchical clustering of patient-specific drug responses (sDSS) with Euclidian distance (cultures and drugs). The heat map is filtered by DSS ≥10 and sDSS ≥ or ≤ 6.5 (n = 74 drugs). The two most sensitive cultures clustered separately and were both of a proneural subtype, with a methylated MGMT promoter. The four least sensitive cultures grouped together in the other major taxonomy; however, among the moderate and least sensitive cultures, no clear pattern was observed in the subtype classification or methylation status of the parent tumor. Even in the cultures clustering together, individual differences in sensitivities to different mechanistic classes of drugs were found (e.g., sensitivity to topoisomerase I inhibitors in T1459 compared to that in T1506, sensitivity to CDK-inhibitors in T1549 compared to that in T1561, sensitivity to mTOR-pathway inhibitors in T1456 compared to that in T1502, and sensitivity to MEK1/2 inhibitors in T1461 compared to that in T1550). Subtype; M: Mesenchymal, PN: proneural, gray box: not available data. MGMT promoter status: ME: Methylated MGMT promoter, UN: Unmethylated MGMT promoter, gray box: not available data
Fig. 5
Fig. 5
Heterogeneity in patient-specific drug responses in treatment-naïve GSCs. (a) Dot plot of the distribution of the patient-specific responses (sDSS) in T1456 to all drugs with DSS ≥10 in any GSC culture displays the enrichment of proteasome inhibitor (green) clustering with increased culture specificity and the insensitivity to aurora pathway inhibitors (yellow). (b) Dot plot displaying the distribution of the drug categories clustering with the highest patient-selectivity in individual GSC cultures. Drugs are filtered by DSS ≥10 and sDSS ≥3, and drug classes are filtered by O/E ≥ 3 for the individual culture. Classes of drugs enriched in individual cultures are highlighted and display the extensive intertumoral heterogeneity in patient-specific vulnerabilities to anticancer drugs. In cultures T1459, T1506 and T1547, the top 20 selective drug responses are presented. Of the drugs with DSS ≥10, three drugs singly target HDAC, whereas two drugs (CUDC-907 and CUDC-101) have dual targets by targeting HDAC along with PI3K or EGFR/Her2, respectively. In T1547, all five drugs that singly or as a dual target inhibit HDAC were found to have the highest patient selectivity and were highlighted within the category of HDAC inhibitors. For the PLK1 inhibitors and bcl-2 inhibitors, O/E was < 3 as only 2 drugs were represented in the drug collection; however, these drugs are highlighted as they displayed unique selectivity in T1459 and T1547, respectively. (c) Dose-response curves of selected drug responses displaying the most sensitive tumor (colored line, drug response is highlighted with enhanced rim in dot plot in B) and the least sensitive tumor (black line) compared to the average response in GBM (dashed line). All drugs have (i) been tested in clinical trials of GBM (nintedanib, paclitaxel, topotecan), (ii) are currently in clinical trials of GBM (belinostat (NCT02137759), sapanisertib (NCT02142803), and selinexor (NCT01986348), clinicaltrials.gov) or (iii) represent drugs within a class that are being investigated in GBM (carfilzomib; proteasome inhibitors, idasanutlin; mdm2 inhibitors, clinicaltrials.gov). Both insensitive and highly sensitive cultures are found in response to each drug

References

    1. Rønning PA, Helseth E, Meling TR, Johannesen TB. A population-based study on the effect of temozolomide in the treatment of glioblastoma multiforme. Neuro-Oncology. 2012;14:1178–1184. doi: 10.1093/neuonc/nos153.
    1. Touat M, Idbaih A, Sanson M, Ligon KL. Glioblastoma targeted therapy: updated approaches from recent biological insights. Ann Oncol. 2017;28:1457–1472. doi: 10.1093/annonc/mdx106.
    1. Brennan CW, Verhaak RGW, McKenna A, Campos B, Noushmehr H, Salama SR, et al. The somatic genomic landscape of glioblastoma. Cell. 2013;155:462–477. doi: 10.1016/j.cell.2013.09.034.
    1. Verhaak RGW, Hoadley KA, Purdom E, Wang V, Qi Y, Wilkerson MD, et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell. 2010;17:98–110. doi: 10.1016/j.ccr.2009.12.020.
    1. Sottoriva A, Spiteri I, Piccirillo SGM, Touloumis A, Collins VP, Marioni JC, et al. Intratumor heterogeneity in human glioblastoma reflects cancer evolutionary dynamics. Proc Natl Acad Sci U S A. 2013;110:4009–4014. doi: 10.1073/pnas.1219747110.
    1. Patel AP, Tirosh I, Trombetta JJ, Shalek AK, Gillespie SM, Wakimoto H, et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science. 2014;344:1396–1401. doi: 10.1126/science.1254257.
    1. Chinot OL, Wick W, Mason W, Henriksson R, Saran F, Nishikawa R, et al. Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N Engl J Med. 2014;370:709–722. doi: 10.1056/NEJMoa1308345.
    1. Westphal M, Heese O, Steinbach JP, Schnell O, Schackert G, Mehdorn M, et al. A randomised, open label phase III trial with nimotuzumab, an anti-epidermal growth factor receptor monoclonal antibody in the treatment of newly diagnosed adult glioblastoma. Eur J Cancer. 2015;51:522–532. doi: 10.1016/j.ejca.2014.12.019.
    1. Stupp R, Taillibert S, Kanner A, Read W, Steinberg DM, Lhermitte B, et al. Effect of tumor-treating fields plus maintenance Temozolomide vs maintenance Temozolomide alone on survival in patients with glioblastoma. JAMA. 2017;318:2306–2311. doi: 10.1001/jama.2017.18718.
    1. De Witt Hamer PC. Small molecule kinase inhibitors in glioblastoma: a systematic review of clinical studies. Neuro-Oncology. 2010;12:304–316. doi: 10.1093/neuonc/nop068.
    1. Sepúlveda-Sánchez JM, Vaz MÁ, Balana C, Gil-Gil M, Reynés G, Gallego Ó, et al. Phase II trial of dacomitinib, a pan-human EGFR tyrosine kinase inhibitor, in recurrent glioblastoma patients with EGFR amplification. Neuro-Oncology. 2017;19:1522–1531. doi: 10.1093/neuonc/nox105.
    1. Lan X, Jörg DJ, Cavalli FMG, Richards LM, Nguyen LV, Vanner RJ, et al. Fate mapping of human glioblastoma reveals an invariant stem cell hierarchy. Nature. 2017;549:227–232. doi: 10.1038/nature23666.
    1. Lee J, Kotliarova S, Kotliarov Y, Li A, Su Q, Donin NM, et al. Tumor stem cells derived from glioblastomas cultured in bFGF and EGF more closely mirror the phenotype and genotype of primary tumors than do serum-cultured cell lines. Cancer Cell. 2006;9:391–403. doi: 10.1016/j.ccr.2006.03.030.
    1. Vik-Mo EO, Sandberg C, Olstorn H, Varghese M, Brandal P, Ramm-Pettersen J, et al. Brain tumor stem cells maintain overall phenotype and tumorigenicity after in vitro culturing in serum-free conditions. Neuro-Oncology. 2010;12:1220–1230. doi: 10.1093/neuonc/noq102.
    1. Davis B, Shen Y, Poon CC, Luchman HA, Stechishin OD, Pontifex CS, et al. Comparative genomic and genetic analysis of glioblastoma-derived brain tumor-initiating cells and their parent tumors. Neuro-Oncology. 2016;18:350–360. doi: 10.1093/neuonc/nov143.
    1. Rosenberg S, Verreault M, Schmitt C, Guegan J, Guehennec J, Levasseur C, et al. Multi-omics analysis of primary glioblastoma cell lines shows recapitulation of pivotal molecular features of parental tumors. Neuro-Oncology. 2017;19:219–228.
    1. Piccirillo SGM, Colman S, Potter NE, van Delft FW, Lillis S, Carnicer M-J, et al. Genetic and functional diversity of propagating cells in glioblastoma. Stem Cell Reports. 2015;4:7–15. doi: 10.1016/j.stemcr.2014.11.003.
    1. Visnyei K, Onodera H, Damoiseaux R, Saigusa K, Petrosyan S, De Vries D, et al. A molecular screening approach to identify and characterize inhibitors of glioblastoma stem cells. Mol Cancer Ther. 2011;10:1818–1828. doi: 10.1158/1535-7163.MCT-11-0268.
    1. Hothi P, Martins TJ, Chen L, Deleyrolle L, Yoon J-G, Reynolds B, et al. High-throughput chemical screens identify disulfiram as an inhibitor of human glioblastoma stem cells. Oncotarget. 2012;3:1124–1136. doi: 10.18632/oncotarget.707.
    1. Lun X, Wells JC, Grinshtein N, King JC, Hao X, Dang N-H, et al. Disulfiram when combined with copper enhances the therapeutic effects of Temozolomide for the treatment of glioblastoma. Clin Cancer Res. 2016;22:3860–3875. doi: 10.1158/1078-0432.CCR-15-1798.
    1. KK-H Y, Taylor JT, Pathmanaban ON, Youshani AS, Beyit D, Dutko-Gwozdz J, et al. High content screening of patient-derived cell lines highlights the potential of non-standard chemotherapeutic agents for the treatment of glioblastoma. PLoS One. 2018;13:e0193694. doi: 10.1371/journal.pone.0193694.
    1. Pemovska T, Kontro M, Yadav B, Edgren H, Eldfors S, Szwajda A, et al. Individualized systems medicine strategy to tailor treatments for patients with chemorefractory acute myeloid leukemia. Cancer Discovery. 2013;3:1416–1429. doi: 10.1158/-13-0350.
    1. Yadav B, Pemovska T, Szwajda A, Kulesskiy E, Kontro M, Karjalainen R, et al. Quantitative scoring of differential drug sensitivity for individually optimized anticancer therapies. Sci Rep. 2014;4:5193. doi: 10.1038/srep05193.
    1. Mao P, Joshi K, Li J, Kim S-H, Li P, Santana-Santos L, et al. Mesenchymal glioma stem cells are maintained by activated glycolytic metabolism involving aldehyde dehydrogenase 1A3. Proc Natl Acad Sci U S A. 2013;110:8644–8649. doi: 10.1073/pnas.1221478110.
    1. Phillips HS, Kharbanda S, Chen R, Forrest WF, Soriano RH, Wu TD, 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. doi: 10.1016/j.ccr.2006.02.019.
    1. Varghese M, Olstorn H, Sandberg C, Vik-Mo EO, Noordhuis P, Nistér M, et al. A comparison between stem cells from the adult human brain and from brain tumors. Neurosurgery. 2008;63:1022–1033. doi: 10.1227/01.NEU.0000335792.85142.B0.
    1. Karajannis MA, Legault G, Fisher MJ, Milla SS, Cohen KJ, Wisoff JH, et al. Phase II study of sorafenib in children with recurrent or progressive low-grade astrocytomas. Neuro-Oncology. 2014;16:1408–1416. doi: 10.1093/neuonc/nou059.
    1. Weller M, Butowski N, Tran DD, Recht LD, Lim M, Hirte H, et al. Rindopepimut with temozolomide for patients with newly diagnosed, EGFRvIII-expressing glioblastoma (ACT IV): a randomised, double-blind, international phase 3 trial. Lancet Oncol. 2017;18:1373–1385. doi: 10.1016/S1470-2045(17)30517-X.
    1. Le Tourneau C, Delord J-P, Gonçalves A, Gavoille C, Dubot C, Isambert N, et al. Molecularly targeted therapy based on tumour molecular profiling versus conventional therapy for advanced cancer (SHIVA): a multicentre, open-label, proof-of-concept, randomised, controlled phase 2 trial. Lancet Oncol. 2015;16:1324–1334. doi: 10.1016/S1470-2045(15)00188-6.
    1. Letai A. Functional precision cancer medicine—moving beyond pure genomics. Nature. 2017;23:1028–1035.
    1. Iwadate Y, Fujimoto S, Namba H, Yamaura A. Promising survival for patients with glioblastoma multiforme treated with individualised chemotherapy based on in vitro drug sensitivity testing. Br J Cancer. 2003;89:1896–1900. doi: 10.1038/sj.bjc.6601376.
    1. Quartararo CE, Reznik E, deCarvalho AC, Mikkelsen T, Stockwell BR. High-throughput screening of patient-derived cultures reveals potential for precision medicine in glioblastoma. ACS Med Chem Lett. 2015;6:948–952. doi: 10.1021/acsmedchemlett.5b00128.
    1. Meyer M, Reimand J, Lan X, Head R, Zhu X, Kushida M, et al. Single cell-derived clonal analysis of human glioblastoma links functional and genomic heterogeneity. Proc Natl Acad Sci U S A. 2015;112:851–856. doi: 10.1073/pnas.1320611111.
    1. Segerman A, Niklasson M, Haglund C, Bergström T, Jarvius M, Xie Y, et al. Clonal variation in drug and radiation response among glioma-initiating cells is linked to proneural- mesenchymal transition. Cell Rep. 2016;17:2994–3009. doi: 10.1016/j.celrep.2016.11.056.
    1. Muhic A, Poulsen HS, Sorensen M, Grunnet K, Lassen U. Phase II open-label study of nintedanib in patients with recurrent glioblastoma multiforme. J Neuro-Oncol. 2013;111:205–212. doi: 10.1007/s11060-012-1009-y.
    1. Chang SM, Kuhn JG, Robins HI, Schold SC, Spence AM, Berger MS, et al. A phase II study of paclitaxel in patients with recurrent malignant glioma using different doses depending upon the concomitant use of anticonvulsants: a north American brain tumor consortium report. Cancer. 2001;91:417–422. doi: 10.1002/1097-0142(20010115)91:2<417::AID-CNCR1016>;2-9.
    1. Wick W, Gorlia T, Bady P, Platten M, van den Bent MJ, Taphoorn MJB, et al. Phase II study of radiotherapy and Temsirolimus versus Radiochemotherapy with Temozolomide in patients with newly diagnosed glioblastoma without MGMT promoter Hypermethylation (EORTC 26082) Clin Cancer Res. 2016;22:4797–4806. doi: 10.1158/1078-0432.CCR-15-3153.
    1. Lesimple T, Riffaud L, Frappaz D, Ben Hassel M, Gédouin D, Bay J-O, et al. Topotecan in combination with radiotherapy in unresectable glioblastoma: a phase 2 study. J Neuro-Oncol. 2009;93:253–260. doi: 10.1007/s11060-008-9774-3.
    1. Blough MD, Westgate MR, Beauchamp D, Kelly JJ, Stechishin O, Ramirez AL, et al. Sensitivity to temozolomide in brain tumor initiating cells. Neuro-Oncology. 2010;12:756–760. doi: 10.1093/neuonc/noq032.
    1. Kierulf-Vieira KS, Sandberg CJ, Grieg Z, Günther C-C, Langmoen IA, Vik-Mo EO. Wnt inhibition is dysregulated in gliomas and its re-establishment inhibits proliferation and tumor sphere formation. Exp. Cell Res. 2015.
    1. Skaga E, Skaga IØ, Grieg Z, Sandberg CJ, Langmoen IA, Vik-Mo EO. The efficacy of a coordinated pharmacological blockade in glioblastoma stem cells with nine repurposed drugs using the CUSP9 strategy. J Cancer Res Clin Oncol. 2019;58:256–213.
    1. Li A, Walling J, Kotliarov Y, Center A, Steed ME, Ahn SJ, et al. Genomic changes and gene expression profiles reveal that established glioma cell lines are poorly representative of primary human gliomas. Mol Cancer Res. 2008;6:21–30. doi: 10.1158/1541-7786.MCR-07-0280.
    1. Vik-Mo EO, Nyakas M, Mikkelsen BV, Moe MC, Due-Tønnesen P, Suso EMI, et al. Therapeutic vaccination against autologous cancer stem cells with mRNA-transfected dendritic cells in patients with glioblastoma. Cancer Immunol Immunother Heidelberg. 2013;62:1499–1509. doi: 10.1007/s00262-013-1453-3.
    1. Sandberg CJ, Altschuler G, Jeong J, Strømme KK, Stangeland B, Murrell W, et al. Comparison of glioma stem cells to neural stem cells from the adult human brain identifies dysregulated Wnt- signaling and a fingerprint associated with clinical outcome. Exp Cell Res. 2013;319:2230–2243. doi: 10.1016/j.yexcr.2013.06.004.
    1. Laks DR, Masterman-Smith M, Visnyei K, Angenieux B, Orozco NM, Foran I, et al. Neurosphere formation is an independent predictor of clinical outcome in malignant glioma. Stem Cells. 2009;27:980–987. doi: 10.1002/stem.15.
    1. D'Alessandris QG, Biffoni M, Martini M, Runci D, Buccarelli M, Cenci T, et al. The clinical value of patient-derived glioblastoma tumorspheres in predicting treatment response. Neuro-Oncology. 2017;19:1097–1108. doi: 10.1093/neuonc/now304.

Source: PubMed

3
Abonnere