Modulations in the Peripheral Immune System of Glioblastoma Patient Is Connected to Therapy and Tumor Progression-A Case Report from the IMMO-GLIO-01 Trial

Paul F Rühle, Nicole Goerig, Roland Wunderlich, Rainer Fietkau, Udo S Gaipl, Annedore Strnad, Benjamin Frey, Paul F Rühle, Nicole Goerig, Roland Wunderlich, Rainer Fietkau, Udo S Gaipl, Annedore Strnad, Benjamin Frey

Abstract

Immune responses are important for efficient tumor elimination, also in immune privileged organs such as the brain. Fostering antitumor immunity has therefore become an important challenge in cancer therapy. This cannot only be achieved by immunotherapies as already standard treatments such as radiotherapy and chemotherapy modify the immune system. Consequently, the understanding of how the tumor, the tumor microenvironment, and immune system are modulated by cancer therapy is required for prognosis, prediction, and therapy adaption. The prospective, explorative, and observational IMMO-GLIO-01 trial was initiated to examine the detailed immune status and its modulation of about 50 patients suffering from primary glioblastoma multiforme (GBM) or anaplastic astrocytoma during standard therapy. Prior to the study, a flow cytometry-based assay was established allowing the analysis of 34 immune cell subsets and their activation state. Here, we present the case of the first and longest accompanied patient, a 53-year-old woman suffering from GBM in the front left lobe. In context of tumor progression and therapy, we describe the modulation of the peripheral immune status over 17 months. Distinct immune modulations that were connected to therapy response or tumor progression were identified. Inter alia, a shift of CD4:CD8 ratio was observed that correlated with tumor progression. Twice we observed a unique composition of peripheral immune cells that correlated with tumor progression. Thus, following up these immune modulations in a closely-meshed manner is of high prognostic and predictive relevance for supporting personalized therapy and increasing therapy success. Clinical Trial registration: ClinicalTrials.gov, identifier NCT02022384 (registered retrospectively on 13th of December, 2013).

Keywords: glioblastoma multiforme; immune status; immunophenotyping; liquid biopsy; personalized medicine; radiochemotherapy.

Figures

Figure 1
Figure 1
Individual therapy schedule and clinical response of the reported patient with glioblastoma. The time line depicts treatment (left side) and clinical response (right side) including pathological abnormalities and tumor progression. All numbers stated are in weeks with zero simultaneously being the starting point of radiochemotherapy and the evaluation of the immune status (indicated by red blood drops). The colors represent treatments or therapy outcomes according to the color code presented in the top right corner. This color code was also used for data presentation in Figure 3. The magnetic resonance imaging scans directly refer to the according image in Figures 2C–G.
Figure 2
Figure 2
Tumor progression identified by consecutive magnetic resonance imaging (MRI) scans reveals transient therapy success and two relapses. MRI scans showing T1- (upper panel) and T2-weighted images (lower panel) of a glioblastoma multiforme in the right temporal lobe, from primary diagnosis through the different stages of tumor progression. After primary diagnosis (A) and subsequent complete resection (B), standard concomitant radiochemotherapy (RCT) followed by adjuvant chemotherapy with Temodal (TMZ) according to Stupp et al. was performed. The first follow-up MRI (3 months after RCT) showed no signs of tumor recurrence (C). Two months later, an MRI was performed due to TMZ cytotoxicity. There were hints of tumor relapse (D), which was histologically confirmed after the lesion had been completely removed by surgery. Yet only 2 (E) and 3 months (F) after this second resection, tumor progression was first suspected and then confirmed. Thus, a conversion of therapy was decided, the therapy of choice being secondary radiotherapy combined with Avastin. This led to tumor regression as observed during the 3-month follow-up during aftercare with Avastin and irinotecan (G).
Figure 3
Figure 3
Multimodal therapy of glioblastoma has high impact on immune cells of the peripheral blood. The graphs of (A) and (C–F) display the absolute cell counts of immune cells or immune cell subsets in the peripheral blood, and in (B), the ratio from CD4+ helper T cells to CD8+ cytotoxic T cells is displayed. All immune cells were determined by a multicolor flow cytometry-based assay and directly detected in whole blood samples. The blood was always drawn prior to administering the drug or radiotherapy on the respective day. The color code represents the last applied therapy or pathological finding (red) and was applied according to Figure 1. The green background marks normal values and the red one deviation from it.

References

    1. Ostrom QT, Gittleman H, Liao P, Rouse C, Chen Y, Dowling J, et al. CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2007–2011. Neuro Oncol (2014) 16(Suppl 4):iv1–63.10.1093/neuonc/nou223
    1. Stupp R, Dietrich PY, Ostermann Kraljevic S, Pica A, Maillard I, Maeder P, et al. Promising survival for patients with newly diagnosed glioblastoma multiforme treated with concomitant radiation plus temozolomide followed by adjuvant temozolomide. J Clin Oncol (2002) 20(5):1375–82.10.1200/JCO.2002.20.5.1375
    1. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med (2005) 352(10):987–96.10.1056/NEJMoa043330
    1. McLendon RE, Halperin EC. Is the long-term survival of patients with intracranial glioblastoma multiforme overstated? Cancer (2003) 98(8):1745–8.10.1002/cncr.11666
    1. Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol (2007) 114(2):97–109.10.1007/s00401-007-0278-6
    1. Fecci PE, Heimberger AB, Sampson JH. Immunotherapy for primary brain tumors: no longer a matter of privilege. Clin Cancer Res (2014) 20(22):5620–9.10.1158/1078-0432.CCR-14-0832
    1. Postow MA, Callahan MK, Barker CA, Yamada Y, Yuan J, Kitano S, et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N Engl J Med (2012) 366(10):925–31.10.1056/NEJMoa1112824
    1. Bielamowicz K, Khawja S, Ahmed N. Adoptive cell therapies for glioblastoma. Front Oncol (2013) 3:275.10.3389/fonc.2013.00275
    1. Ransohoff RM, Kivisakk P, Kidd G. Three or more routes for leukocyte migration into the central nervous system. Nat Rev Immunol (2003) 3(7):569–81.10.1038/nri1130
    1. Van Woensel M, Mathivet T, Wauthoz N, Rosiere R, Garg AD, Agostinis P, et al. Sensitization of glioblastoma tumor micro-environment to chemo- and immunotherapy by galectin-1 intranasal knock-down strategy. Sci Rep (2017) 7(1):1217.10.1038/s41598-017-01279-1
    1. Eder K, Kalman B. The dynamics of interactions among immune and glioblastoma cells. Neuromolecular Med (2015) 17(4):335–52.10.1007/s12017-015-8362-x
    1. Adamczyk LA, Williams H, Frankow A, Ellis HP, Haynes HR, Perks C, et al. Current understanding of circulating tumor cells – potential value in malignancies of the central nervous system. Front Neurol (2015) 6:174.10.3389/fneur.2015.00174
    1. Ruhle PF, Fietkau R, Gaipl US, Frey B. Development of a modular assay for detailed immunophenotyping of peripheral human whole blood samples by multicolor flow cytometry. Int J Mol Sci (2016) 17(8):1–29.10.3390/ijms17081316
    1. Frey B, Rubner Y, Wunderlich R, Weiss EM, Pockley AG, Fietkau R, et al. Induction of abscopal anti-tumor immunity and immunogenic tumor cell death by ionizing irradiation – implications for cancer therapies. Curr Med Chem (2012) 19(12):1751–64.10.2174/092986712800099811
    1. Minniti G, Lanzetta G, Scaringi C, Caporello P, Salvati M, Arcella A, et al. Phase II study of short-course radiotherapy plus concomitant and adjuvant temozolomide in elderly patients with glioblastoma. Int J Radiat Oncol Biol Phys (2012) 83(1):93–9.10.1016/j.ijrobp.2011.06.1992
    1. Belka C, Ottinger H, Kreuzfelder E, Weinmann M, Lindemann M, Lepple-Wienhues A, et al. Impact of localized radiotherapy on blood immune cells counts and function in humans. Radiother Oncol (1999) 50(2):199–204.10.1016/S0167-8140(98)00130-3
    1. Ostermann S, Csajka C, Buclin T, Leyvraz S, Lejeune F, Decosterd LA, et al. Plasma and cerebrospinal fluid population pharmacokinetics of temozolomide in malignant glioma patients. Clin Cancer Res (2004) 10(11):3728–36.10.1158/1078-0432.CCR-03-0807
    1. Hirose Y, Berger MS, Pieper RO. p53 effects both the duration of G2/M arrest and the fate of temozolomide-treated human glioblastoma cells. Cancer Res (2001) 61(5):1957–63.
    1. Hickey MJ, Malone CC, Erickson KE, Gomez GG, Young EL, Liau LM, et al. Implementing preclinical study findings to protocol design: translational studies with alloreactive CTL for gliomas. Am J Transl Res (2012) 4(1):114–26.
    1. Grossman SA, Ye X, Lesser G, Sloan A, Carraway H, Desideri S, et al. Immunosuppression in patients with high-grade gliomas treated with radiation and temozolomide. Clin Cancer Res (2011) 17(16):5473–80.10.1158/1078-0432.CCR-11-0774
    1. Lotfi R, Lee JJ, Lotze MT. Eosinophilic granulocytes and damage-associated molecular pattern molecules (DAMPs): role in the inflammatory response within tumors. J Immunother (2007) 30(1):16–28.10.1097/01.cji.0000211324.53396.f6
    1. Davis BP, Rothenberg ME. Eosinophils and cancer. Cancer Immunol Res (2014) 2(1):1–8.10.1158/2326-6066.CIR-13-0196
    1. Gatault S, Legrand F, Delbeke M, Loiseau S, Capron M. Involvement of eosinophils in the anti-tumor response. Cancer Immunol Immunother (2012) 61(9):1527–34.10.1007/s00262-012-1288-3
    1. Carretero R, Sektioglu IM, Garbi N, Salgado OC, Beckhove P, Hammerling GJ. Eosinophils orchestrate cancer rejection by normalizing tumor vessels and enhancing infiltration of CD8(+) T cells. Nat Immunol (2015) 16(6):609–17.10.1038/ni.3159
    1. Lauber K, Ernst A, Orth M, Herrmann M, Belka C. Dying cell clearance and its impact on the outcome of tumor radiotherapy. Front Oncol (2012) 2:116.10.3389/fonc.2012.00116
    1. Kotlan B, Simsa P, Teillaud JL, Fridman WH, Toth J, McKnight M, et al. Novel ganglioside antigen identified by B cells in human medullary breast carcinomas: the proof of principle concerning the tumor-infiltrating B lymphocytes. J Immunol (2005) 175(4):2278–85.10.4049/jimmunol.175.4.2278
    1. Nzula S, Going JJ, Stott DI. Antigen-driven clonal proliferation, somatic hypermutation, and selection of B lymphocytes infiltrating human ductal breast carcinomas. Cancer Res (2003) 63(12):3275–80.
    1. Dong HP, Elstrand MB, Holth A, Silins I, Berner A, Trope CG, et al. NK- and B-cell infiltration correlates with worse outcome in metastatic ovarian carcinoma. Am J Clin Pathol (2006) 125(3):451–8.10.1309/15B66DQMFYYM78CJ
    1. Tsou P, Katayama H, Ostrin EJ, Hanash SM. The emerging role of B cells in tumor immunity. Cancer Res (2016) 76(19):5597–601.10.1158/0008-5472.CAN-16-0431
    1. Rubtsov AV, Rubtsova K, Kappler JW, Jacobelli J, Friedman RS, Marrack P. CD11c-expressing B cells are located at the T cell/B cell border in spleen and are potent APCs. J Immunol (2015) 195(1):71–9.10.4049/jimmunol.1500055
    1. Inoue S, Leitner WW, Golding B, Scott D. Inhibitory effects of B cells on antitumor immunity. Cancer Res (2006) 66(15):7741–7.10.1158/0008-5472.CAN-05-3766
    1. Shalapour S, Font-Burgada J, Di Caro G, Zhong Z, Sanchez-Lopez E, Dhar D, et al. Immunosuppressive plasma cells impede T-cell-dependent immunogenic chemotherapy. Nature (2015) 521(7550):94–8.10.1038/nature14395
    1. Lampson LA. Monoclonal antibodies in neuro-oncology: getting past the blood-brain barrier. MAbs (2011) 3(2):153–60.10.4161/mabs.3.2.14239
    1. Verhoeff JJ, van Tellingen O, Claes A, Stalpers LJ, van Linde ME, Richel DJ, et al. Concerns about anti-angiogenic treatment in patients with glioblastoma multiforme. BMC Cancer (2009) 9:444.10.1186/1471-2407-9-444
    1. Goel S, Duda DG, Xu L, Munn LL, Boucher Y, Fukumura D, et al. Normalization of the vasculature for treatment of cancer and other diseases. Physiol Rev (2011) 91(3):1071–121.10.1152/physrev.00038.2010
    1. Dirkx AE, oude Egbrink MG, Castermans K, van der Schaft DW, Thijssen VL, Dings RP, et al. Anti-angiogenesis therapy can overcome endothelial cell anergy and promote leukocyte-endothelium interactions and infiltration in tumors. FASEB J (2006) 20(6):621–30.10.1096/fj.05-4493com
    1. Osada T, Chong G, Tansik R, Hong T, Spector N, Kumar R, et al. The effect of anti-VEGF therapy on immature myeloid cell and dendritic cells in cancer patients. Cancer Immunol Immunother (2008) 57(8):1115–24.10.1007/s00262-007-0441-x
    1. Xu Y, Villalona-Calero MA. Irinotecan: mechanisms of tumor resistance and novel strategies for modulating its activity. Ann Oncol (2002) 13(12):1841–51.10.1093/annonc/mdf337
    1. Kawahara M. Irinotecan in the treatment of small cell lung cancer: a review of patient safety considerations. Expert Opin Drug Saf (2006) 5(2):303–12.10.1517/14740338.5.2.303
    1. Faget J, Bendriss-Vermare N, Gobert M, Durand I, Olive D, Biota C, et al. ICOS-ligand expression on plasmacytoid dendritic cells supports breast cancer progression by promoting the accumulation of immunosuppressive CD4+ T cells. Cancer Res (2012) 72(23):6130–41.10.1158/0008-5472.CAN-12-2409
    1. Jensen TO, Schmidt H, Moller HJ, Donskov F, Hoyer M, Sjoegren P, et al. Intratumoral neutrophils and plasmacytoid dendritic cells indicate poor prognosis and are associated with pSTAT3 expression in AJCC stage I/II melanoma. Cancer (2012) 118(9):2476–85.10.1002/cncr.26511
    1. Labidi-Galy SI, Treilleux I, Goddard-Leon S, Combes JD, Blay JY, Ray-Coquard I, et al. Plasmacytoid dendritic cells infiltrating ovarian cancer are associated with poor prognosis. Oncoimmunology (2012) 1(3):380–2.10.4161/onci.18801
    1. Beckebaum S, Zhang X, Chen X, Yu Z, Frilling A, Dworacki G, et al. Increased levels of interleukin-10 in serum from patients with hepatocellular carcinoma correlate with profound numerical deficiencies and immature phenotype of circulating dendritic cell subsets. Clin Cancer Res (2004) 10(21):7260–9.10.1158/1078-0432.CCR-04-0872
    1. Treilleux I, Blay JY, Bendriss-Vermare N, Ray-Coquard I, Bachelot T, Guastalla JP, et al. Dendritic cell infiltration and prognosis of early stage breast cancer. Clin Cancer Res (2004) 10(22):7466–74.10.1158/1078-0432.CCR-04-0684
    1. Vermi W, Bonecchi R, Facchetti F, Bianchi D, Sozzani S, Festa S, et al. Recruitment of immature plasmacytoid dendritic cells (plasmacytoid monocytes) and myeloid dendritic cells in primary cutaneous melanomas. J Pathol (2003) 200(2):255–68.10.1002/path.1344
    1. Le Mercier I, Poujol D, Sanlaville A, Sisirak V, Gobert M, Durand I, et al. Tumor promotion by intratumoral plasmacytoid dendritic cells is reversed by TLR7 ligand treatment. Cancer Res (2013) 73(15):4629–40.10.1158/0008-5472.CAN-12-3058
    1. Preynat-Seauve O, Schuler P, Contassot E, Beermann F, Huard B, French LE. Tumor-infiltrating dendritic cells are potent antigen-presenting cells able to activate T cells and mediate tumor rejection. J Immunol (2006) 176(1):61–7.10.4049/jimmunol.176.1.61
    1. Liu C, Lou Y, Lizee G, Qin H, Liu S, Rabinovich B, et al. Plasmacytoid dendritic cells induce NK cell-dependent, tumor antigen-specific T cell cross-priming and tumor regression in mice. J Clin Invest (2008) 118(3):1165–75.10.1172/JCI33583
    1. Quandt D, Zucht HD, Amann A, Wulf-Goldenberg A, Borrebaeck C, Cannarile M, et al. Implementing liquid biopsies into clinical decision making for cancer immunotherapy. Oncotarget (2017).10.18632/oncotarget.17397
    1. Burdach SE, Bliesener JA, Evers KG, Gharib M. [Pre- and retrorenal neuroblastoma without kidney displacement]. Klin Padiatr (1983) 195(1):52–6.10.1055/s-2008-1034041

Source: PubMed

3
订阅