Predictors of Response to Autologous Dendritic Cell Therapy in Glioblastoma Multiforme

Chia-Ing Jan, Wan-Chen Tsai, Horng-Jyh Harn, Woei-Cherng Shyu, Ming-Chao Liu, Hsin-Man Lu, Shao-Chih Chiu, Der-Yang Cho, Chia-Ing Jan, Wan-Chen Tsai, Horng-Jyh Harn, Woei-Cherng Shyu, Ming-Chao Liu, Hsin-Man Lu, Shao-Chih Chiu, Der-Yang Cho

Abstract

Background: Glioblastoma (GBM) is the most common and lethal primary malignant glioma in adults. Dendritic cell (DC) vaccines have demonstrated promising results in GBM clinical trials. However, some patients do not respond well to DC therapy, with survival rates similar to those of conventional therapy. We retrospectively analyzed clinical and laboratory data to evaluate the factors affecting vaccine treatment.

Methods: Forty-seven patients with de novo GBM were enrolled at China Medical University Hospital between 2005 and 2010 and divided into two subgroups. One subgroup of 27 patients received postsurgical adjuvant immunotherapy with autologous dendritic cell/tumor antigen vaccine (ADCTA) in conjunction with conventional treatment of concomitant chemoradiotherapy (CCRT) with temozolomide. The other 20 patients received only postsurgical conventional treatment without immunotherapy. Immunohistochemistry for CD45, CD4, CD8, programed death ligand 1 (PD-L1), and programed death 1 (PD-1) was performed on sections of surgical tumor specimens and peripheral blood mononuclear cells (PBMCs). Pearson's correlation, Cox proportional hazard model, and Kaplan-Meier analyses were performed to examine the correlations between the prognostic factors and survival rates.

Results: Younger age (<57 years), gross total resection, and CCRT and PD-1+ lymphocyte counts were significant prognostic factors of overall survival (OS) and progression-free survival (PFS) in the ADCTA group. Sex, CD45+ lymphocyte count, CD4+ or CD8+ lymphocyte count, tumor PD-L1 expression, isocitrate dehydrogenase 1 mutation, and O6 methylguanine-DNA methyltransferase promoter methylation status were not significant factors in both groups. In the ADCTA group, patients with tumor-infiltrating lymphocytes (TILs) with a lower PD-1+/CD8+ ratio (≤0.21) had longer OS and PFS (median OS 60.97 months, P < 0.001 and PFS 11.2 months, P < 0.008) compared to those with higher PD-1+/CD8+ ratio (>0.21) (median OS 20.07 months, P < 0.001 and PFS 4.43 months, P < 0.008). Similar results were observed in patients' PBMCs; lymphocyte counts with lower PD-1+/CD8+ ratio (≤0.197) had longer OS and PFS. There was a significant correlation of PD-1+/CD8+ ratio between TILs and PBMCs (Pearson's correlation R2 = 0.6002, P < 0.001). By contrast, CD4-, CD8-, but PD-1+, CD45+ tumor-infiltrating lymphocytes have no impact on OS and PFS (P = 0.073 and P = 0.249, respectively).

Conclusion: For patients receiving DC vaccine adjuvant therapy, better outcomes are predicted in patients with younger age, with TILs or PBMCs with lower PD-1+/CD8+ ratio, with gross tumor resection, and receiving CCRT.

Keywords: PD-1+/CD8+ ratio; autologous dendritic cell/tumor antigen; cytotoxic T-lymphocytes (CD8+); glioblastoma multiforme; immune checkpoints; peripheral blood mononuclear cell; programmed death protein 1 (PD-1+); tumor-infiltrating lymphocytes.

Figures

Figure 1
Figure 1
Treatment schema and vaccine preparation. Clinical schematic diagram (A). Subjects with primary GBM will be consent for operation and concomitant chemoradiotherapy (CCRT). Subjects assigned to the ADCTA group will be designated to receive dendritic cell (DC) vaccination ten times following the clinical trial schedule after operation. V: visits to hospital, numbers following indicates times of visit. DC vaccine manufacturing protocol (B). In China Medical University Hospital, the DC vaccine is produced in laboratories that meet the requirements of Good Tissue Practices and Good Manufacturing Practices. The final product is used in a clinical trial of autologous DC therapy for GBM patients between years 2005 and 2010.
Figure 2
Figure 2
Immunohistochemistry staining of CD4, CD8, programed death 1 (PD-1), and CD45 expression in tumor-infiltrating lymphocytes in glioblastoma (GBM) tissue and peripheral blood mononuclear cells. Representative photomicrographs showing staining for CD4 (A), CD8 (B), PD-1 (C), and CD45 (D) in GBM histological tissue sections (400× magnification) and CD4 (E), CD8 (F), PD-1 (G), and CD45 (H) in peripheral blood mononuclear cell cytoblocks (400× magnification).
Figure 3
Figure 3
Immunohistochemistry staining pattern of positive and negative PD-L1 expression in GBM tumor cells. Representative photomicrographs showing diffuse strong cell membrane staining for PD-L1 (A), and complete negative staining for PD-L1 (B) in GBM histological tissue sections (400× magnification).
Figure 4
Figure 4
Overall survival (OS) in ADCTA versus reference group glioblastoma patients. Kaplan–Meier survival plots of the OS of 27 ADCTA patients compared to 20 reference patients.
Figure 5
Figure 5
Overall survival (OS) (A) and progression-free survival (PFS) (B) by high or low PD-1+/CD8+ ratio tumor-infiltrating lymphocytes in the ADCTA group. Kaplan–Meier survival plots of OS and PFS of 14 patients with high PD-1+/CD8+ ratio compared to 13 patients with low PD-1+/CD8+ ratio patients.
Figure 6
Figure 6
PD-1+/CD8+ ratios of individual patients’ tumor-infiltrating lymphocytes and overall survival (OS) and progression-free survival (PFS) in the ADCTA group. Pearson’s correlation of each patient’s PD1+/CD8+ ratio value and their OS (A) and PFS time (B) after natural logarithm transformation.
Figure 7
Figure 7
Correlation between tumor-infiltrating lymphocytes and peripheral blood mononuclear cells (PBMCs) in the ADCTA group. Pearson’s correlation between tumor-infiltrating lymphocytes and PBMCs from ADCTA patients (18 patients).
Figure 8
Figure 8
Overall survival (OS) (A) and progression-free survival (PFS) (B) according to high or low PD-1+/CD8+ ratio in peripheral blood mononuclear cells in the ADCTA group. Kaplan–Meier survival plots of OS and PFS of 10 high PD-1+/CD8+ ratio patients compared to 8 low PD-1+/CD8+ ratio patients.
Figure 9
Figure 9
Co-expression of programed death 1 (PD-1) and CD8 in PD-1-positive and CD8-positive tumor-infiltrating lymphocytes and peripheral blood mononuclear cells (PBMCs). Paired GBM tissue sections and PBMC cytoblocks from a representative patient with a high PD-1+/CD8+ ratio were selected for immunofluorescence (IF) analysis of PD-1 and CD8 co-expression. CD8 and PD-1 expression in the TIL of GBM tissue [(A), upper panel] and reconstruction of single slices of the xz and the yz planes in the z-axis stacked image [(A), lower panel]. CD8 and PD-1 expression in PBMCs derived from the same patient [(B), upper panel] and reconstruction of the same immunofluorescent slide [(B), lower panel]. Paired GBM tissue sections and PBMC cytoblocks from a representative patient with a low PD-1+/CD8+ ratio were selected for IF analysis of PD-1 and CD8 co-expression. CD8 and PD-1 expression in the TIL of GBM tissue [(C), upper panel] and reconstruction of single slices of the xz and yz planes in the z-axis stacked image [(C), lower panel]. CD8 and PD-1 expression in PBMCs derived from the same patient [(D), upper panel] and reconstruction of the same immunofluorescent slide [(D), lower panel].
Figure 10
Figure 10
Flow cytometry of peripheral blood mononuclear cells (PBMCs) of representative patients. PBMCs (1 × 105) were collected and stained with antibodies against programed death 1 (PD-1), CD8, and CD3. The percpCy5.5-labeled CD3+ T cells (middle panel) were gated form the size and granularity dot-plot (left panel), then these CD3+ T cells were analyzed by the PE-Cy7 and FITC dot-plot to identify the populations of CD8 and PD-1-expressing cells. Patient 21 had a low PD-1+/CD8+ ratio in the PBMC cytoblock (A). Patient 41 was with a high PD-1+/CD8+ ratio in PBMC cytoblock (B).
Figure 11
Figure 11
Schematic representation of dendritic cell vaccine treatment PD-1+ or PD-1− cytotoxic T cells in the peripheral blood arrive at lymph nodes for antigen activation after vaccination. After reaching the tumor microenvironment, the efficiency of tumor cell killing by cytotoxic T cells is determined by the proportion of PD-1+ cytotoxic T cells.

References

    1. Kleihues P, Louis DN, Scheithauer BW, Rorke LB, Reifenberger G, Burger PC, et al. The WHO classification of tumors of the nervous system. J Neuropathol Exp Neurol (2002) 61:215–25.10.1093/jnen/61.3.215
    1. Wen PY, Kesari S. Malignant glioma in adults. N Engl J Med (2008) 359:492–507.10.1056/NEJMra0708126
    1. Jovcevska I, Kocevar N, Komel R. Glioma and glioblastoma – how much do we (not) know. Mol Clin Oncol (2013) 1:935–41.10.3892/mco.2013.172
    1. Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC, et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomized phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol (2009) 10:459–66.10.1016/S1470-2045(09)70025-7
    1. Park JK, Hodges T, Arko L, Shen M, Dello Iacono D, McNabb A, et al. Scale to predict survival after surgery for recurrent glioblastoma multiforme. J Clin Oncol (2010) 28:3838–43.10.1200/JCO.2010.30.0582
    1. Gilbert MR, Dignam JJ, Armstrong TS, Wefel JS, Blumenthal DT, Vogelbaum MA, et al. A randomized trial of bevacizumab for newly diagnosed glioblastoma. N Engl J Med (2014) 370:699–708.10.1056/NEJMoa1308573
    1. Omuro A, Beal K, Gutin P, Karimi S, Correa DD, Kaley TJ, et al. Phase II study of bevacizumab, temozolomide, and hypofractionated stereotactic radiotherapy for newly diagnosed glioblastoma. Clin Cancer Res (2014) 20:5023–31.10.1158/1078-0432.CCR-14-0822
    1. Lombardi G, Pace A, Pasqualetti F, Rizzato S, Faedi M, Anghileri E, et al. Predictors of survival and effect of short (40 Gy) or standard-course (60 Gy) irradiation plus concomitant temozolomide in elderly patients with glioblastoma: a multicenter retrospective study of AINO (Italian association of neuro-oncology). J Neurooncol (2015) 125:359–67.10.1007/s11060-015-1923-x
    1. Mehta M, Wen P, Nishikawa R, Reardon D, Peters K. Critical review of the addition of tumor treating fields (TTFields) to the existing standard of care for newly diagnosed glioblastoma patients. Crit Rev Oncol Hematol (2017) 111:60–5.10.1016/j.critrevonc.2017.01.005
    1. Gramatzki D, Kickingereder P, Hentschel B, Felsberg J, Herrlinger U, Schackert G. Limited role for extended maintenance temozolomide for newly diagnosed glioblastoma. Neurology (2017) 88:1422–30.10.1212/WNL.0000000000003809
    1. Laws ER, Parney IF, Huang W, Anderson F, Morris AM, Asher A, et al. Survival following surgery and prognostic factors for recently diagnosed malignant glioma: data from the glioma outcomes project. J Neurosurg (2003) 99:467–73.10.3171/jns.2003.99.3.0467
    1. Anguille S, Smits EL, Lion E, van Tendeloo VF, Berneman ZN. Clinical use of dendritic cells for cancer therapy. Lancet Oncol (2014) 15:e257–67.10.1016/S1470-2045(13)70585-0
    1. Wang X, Zhao HY, Zhang FC, Sun Y, Xiong ZY, Jiang XB. Dendritic cell-based vaccine for the treatment of malignant glioma: a systemic review. Cancer Invest (2014) 32:451–7.10.3109/07357907.2014.958234
    1. Yang L, Guo G, Niu XY, Liu J. Dendritic cell-based immunotherapy treatment for glioblastoma multiform. Biomed Res Int (2015) 2015:717530.10.1155/2015/717530
    1. Mallick S, Gandhi AK, Rath GK. Therapeutic approach beyond conventional temozolomide for newly diagnosed glioblastoma: review of the present evidence and future direction. Indian J Med Paediatr Oncol (2015) 36:229–37.10.4103/0971-5851.171543
    1. Sabado RL, Balan S, Bhardwaj N. Dendritic cell-based immunotherapy. Cell Res (2017) 27:74–95.10.1038/cr.2016.157
    1. Xu LW, Chow KK, Lim M, Li G. Current vaccine trials in glioblastoma: a review. J Immunol Res (2014) 2014:796856.10.1155/2014/796856
    1. Van Gool S, Maes W, Ardon H, Verschuere T, Van Cauter S, De Vleeschouwer S. Dendritic cell therapy of high-grade gliomas. Brain Pathol (2009) 19:694–712.10.1111/j.1750-3639.2009.00316.x
    1. Van Gool SW. Brain tumor immunotherapy: what have we learned so far? Front Oncol (2015) 5:98.10.3389/fonc.2015.00098
    1. Yamanaka R, Abe T, Yajima N, Tsuchiya N, Homma J, Kobayashi T, et al. Vaccination of recurrent glioma patients with tumour lysate-pulsed dendritic cells elicits immune responses: results of a clinical phase I/II trial. Br J Cancer (2003) 89:11729.10.1038/sj.bjc.6601268
    1. De Vleeschouwer S, Fieuws S, Rutkowski S, Van Calenbergh F, Van Loon J, Goffin J, et al. Postoperative adjuvant dendritic cell-based immunotherapy in patients with relapsed glioblastoma multiforme. Clin Cancer Res (2008) 14:3098–104.10.1158/1078-0432.CCR-07-4875
    1. Bregy A, Wong TM, Shah AH, Goldberg JM, Komotar RJ. Active immunotherapy using dendritic cells in the treatment of glioblastoma multiforme. Cancer Treat Rev (2013) 39:891–907.10.1016/j.ctrv.2013.05.007
    1. Chang CN, Huang YC, Yang DM, Kikuta K, Wei KJ, Kubota T, et al. A phase I/II clinical trial investigating the adverse and therapeutic effects of a postoperative autologous dendritic cell tumor vaccine in patients with malignant glioma. J Clin Neurosci (2011) 18:1048–54.10.1016/j.jocn.2010.11.034
    1. Cho DY, Yang WK, Lee HC, Hsu DM, Lin HL, Lin SZ, et al. Adjuvant immunotherapy with whole-cell lysate dendritic cells vaccine for glioblastoma multiforme: a phase II clinical trial. World Neurosurg (2012) 77:736–44.10.1016/j.wneu.2011.08.020
    1. Prins RM, Wang X, Soto H, Young E, Lisiero DN, Frong B, et al. Comparison of glioma-associated antigen peptide-loaded versus autologous tumor lysate-loaded dendritic cell vaccination in malignant glioma patients. J Immunother (2013) 36:152–7.10.1097/CJI.0b013e3182811ae4
    1. Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med (2010) 363:711–23.10.1056/NEJMoa1003466
    1. Zhang Y, Huang SD, Gong DJ, Qin YH, Shen QA. Programmed death-1 upregulation is correlated with dysfunction of tumor-infiltrating CD8(+) T lymphocytes in human non-small cell lung cancer. Cell Mol Immunol (2010) 7:389–95.10.1038/cmi.2010.28
    1. Duraiswamy J, Freeman GJ, Coukos G. Therapeutic PD-1 pathway blockade augments with other modalities of immunotherapy T-cell function to prevent immune decline in ovarian cancer. Cancer Res (2013) 73:6900–12.10.1158/0008-5472.CAN-13-1550
    1. Sznol M, Chen L. Antagonist antibodies to PD-1 and B7-H1 (PD-L1) in the treatment of advanced human cancer. Clin Cancer Res (2013) 19:1021–34.10.1158/1078-0432.CCR-12-2063
    1. Berghoff AS, Ricken G, Widhalm G, Rajky O, Hainfellner JA, Birner P, et al. PD1 (CD279) and PD-L1 (CD274, B7H1) expression in primary central nervous system lymphomas (PCNSL). Clin Neuropathol (2014) 33:42–9.10.5414/NP300698
    1. Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med (2015) 372:2018–28.10.1056/NEJMoa1501824
    1. Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science (2015) 348:124–8.10.1126/science.aaa1348
    1. Topalian SL, Taube JM, Anders RA, Pardoll DM. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer (2016) 16:275–87.10.1038/nrc.2016.36
    1. Castro MG, Baker GJ, Lowenstein PR. Blocking immunosuppressive checkpoints for glioma therapy: the more the merrier! Clin Cancer Res (2014) 20:5147–9.10.1158/1078-0432.CCR-14-0820
    1. Vlahovic G, Fecci PE, Reardon D, Sampson JH. Programmed death ligand 1 (PD-L1) as an immunotherapy target in patients with glioblastoma. Neuro Oncol (2015) 17:1043–5.10.1093/neuonc/nov071
    1. Bouffet E, Larouche V, Campbell BB, Merico D, de Borja R, Aronson M, et al. Immune checkpoint inhibition for hypermutant glioblastoma multiforme resulting from germline biallelic mismatch repair deficiency. J Clin Oncol (2016) 34:2206–11.10.1200/JCO.2016.66.6552
    1. Blumenthal DT, Yalon M, Vainer GW, Lossos A, Yust S, Tzach L, et al. Pembrolizumab: first experience with recurrent primary central nervous system (CNS) tumors. J Neurooncol (2016) 129:453–60.10.1007/s11060-016-2190-1
    1. Kohrt HE, Tumeh PC, Benson D, Bhardwaj N, Brody J, Formenti S, et al. Unodynamics: a cancer immunotherapy trials network review of immune monitoring in immuno-oncology clinical trials. J Immunother Cancer (2016) 4:15.10.1186/s40425-016-0118-0
    1. McDermott DF, Atkins MB. PD-1 as a potential target in cancer therapy. Cancer Med (2013) 2:662–73.10.1002/cam4.106
    1. Ahn BJ, Pollack IF, Okada H. Immune-checkpoint blockade and active immunotherapy for glioma. Cancers (Basel) (2013) 5:1379–412.10.3390/cancers5041379
    1. Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, et al. Safety and tumor responses with lambrolizumab (anti-PD1) in melanoma. N Engl J Med (2013) 369:134–44.10.1056/NEJMoa1305133
    1. Shi L, Chen S, Yang L, Li Y. The role of PD-1 and PD-L1 in T-cell immune suppression in patients with hematological malignancies. J Hematol Oncol (2013) 6:74.10.1186/1756-8722-6-74
    1. Cojocaru G, Rotman G, Levy O, Toporik A, Dassa L, Vaknin I, et al. S9. Proffered paper: identification of novel immune checkpoints as targets for cancer immunotherapy. J Immunother Cancer (2014) 2:I5.10.1186/2051-1426-2-S2-I5
    1. Antonios JP, Soto H, Everson RG, Orpilla J, Moughon D, Shin N, et al. PD-1 blockade enhances the vaccination-induced immune response in glioma. JCI Insight (2016) 1(10):e87059.10.1172/jci.insight.87059
    1. Antonios JP, Soto H, Everson RG, Moughon D, Orpilla JR, Shin NP, et al. Immunosuppressive tumor-infiltrating myeloid cells mediate adaptive immune resistance via a PD-1/PD-L1 mechanism in glioblastoma. Neuro Oncol (2017) 19(6):796–807.10.1093/neuonc/now287
    1. Stupp R, Brada M, van den Bent MJ, Tonn JC, Pentheroudakis G. High-grade glioma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol (2014) 3:iii93–101.10.1093/annonc/mdu050
    1. Choi SJ, Choi YI, Kim L, Park IS, Han JY, Kim JM, et al. Preparation of compact agarose cell blocks from the residues of liquid-based cytology samples. Korean J Pathol (2014) 48:351–60.10.4132/KoreanJPathol.2014.48.5.351
    1. Berghoff AS, Kiesel B, Widhalm G, Rajky O, Ricken G, Wöhrer A, et al. Programmed death ligand 1 expression and tumor-infiltrating lymphocytes in glioblastoma. Neuro Oncol (2015) 17:1064–75.10.1093/neuonc/nou307
    1. Nduom EK, Wei J, Yaghi NK, Huang N, Kong LY, Gabrusiewicz K, et al. PD-L1 expression and prognostic impact in glioblastoma. Neuro Oncol (2016) 18:195–205.10.1093/neuonc/nov172
    1. Preusser M, Wöhrer A, Stary S, Höftberger R, Streubel B, Hainfellner JA. Value and limitations of immunohistochemistry and gene sequencing for detection of the IHD1-R132H mutation in diffuse glioma biopsy specimens. J Neuropathol Exp Neurol (2011) 70:715–23.10.1097/NEN.0b013e31822713f0
    1. Agarwal S, Sharma MC, Jha P, Pathak P, Suri V, Sarkar C, et al. Comparative study of IDH1 mutations in gliomas by immunohistochemistry and DNA sequencing. Neuro Oncol (2013) 15:718–26.10.1093/neuonc/not015
    1. Matthew SW, Bill HD, Hai Y. Isocitrate dehydrogenase mutations in gliomas. Neuro Oncol (2016) 18:16–26.10.1093/neuonc/nov136
    1. Tsai KK, Zarzoso I, Daud AI. PD-1 and PD-L1 antibodies for melanoma. Hum Vaccin Immunother (2014) 10:3111–6.10.4161/21645515.2014.983409
    1. Ahmadzadeh M, Johnson LA, Heemskerk B, Wunderlich JR, Dudley ME, White DE, et al. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood (2009) 114:1537–44.10.1182/blood-2008-12-195792
    1. Liu Y, Zeng B, Zhang Z, Zhang Y, Yang R. B7-H1 on myeloid-derived suppressor cells in immune suppression by a mouse model of ovarian cancer. Clin Immunol (2008) 129:471–81.10.1016/j.clim.2008.07.030
    1. Mirghorbani M, Van Gool S, Rezaei N. Myeloid-derived suppressor cells in glioma. Expert Rev Neurother (2013) 13:1395–406.10.1586/14737175.2013.857603
    1. Hosseini M, Yousefifard M, Aziznejad H, Nasirinezhad F. The effect of bone marrow-derived mesenchymal stem cell transplantation on allodynia and hyperalgesia in neuropathic animals: a systematic review with meta-analysis. Biol Blood Marrow Transplant (2015) 21:1537–44.10.1016/j.bbmt.2015.05.008
    1. Heiland DH, Haaker G, Delev D, Mercas B, Masalha W, Heynckes S, et al. Comprehensive analysis of PD-L1 expression in glioblastoma multiforme. Oncotarget (2017) 8:42214–25.10.18632/oncotarget.15031
    1. Liau LM, Prins RM, Kiertscher SM, Odesa SK, Kremen TJ, Giovannone AJ, et al. Dendritic cell vaccination in glioblastoma patients induces systemic and intracranial T-cell responses modulated by the local central nervous system tumor microenvironment. Clin Cancer Res (2005) 11:5515–25.10.1158/1078-0432.CCR-05-0464
    1. Topalian SL, Drake CG, Pardoll DM. Targeting the PD-1/B7-H1(PD-L1) pathway to activate anti-tumor immunity. Curr Opin Immunol (2012) 24:207–12.10.1016/j.coi.2011.12.009
    1. Sun S, Fei X, Mao Y, Wang X, Garfield DH, Huang O, et al. PD-1(+) immune cell infiltration inversely correlates with survival of operable breast cancer patients. Cancer Immunol Immunother (2014) 63:395–406.10.1007/s00262-014-1519-x
    1. Wei B, Wang L, Zhao XL, Du C, Guo YC, Sun ZG. The upregulation of programmed death 1 on peripheral blood T cells of glioma is correlated with disease progression. Tumour Biol (2014) 35:2923–9.10.1007/s13277-013-1376-9
    1. Zheng P, Zhou Z. Human cancer immunotherapy with PD-1/PD-L1 blockade. Biomark Cancer (2015) 7:15–8.10.4137/BIC.S29325
    1. Razavi SM, Lee KE, Jin BE, Aujla PS, Gholamin S, Li G. Immune evasion strategies of glioblastoma. Front Surg (2016) 3:11.10.3389/fsurg.2016.00011
    1. Keles GE, Anderson B, Berger MS. The effect of extent of resection on time to tumor progression and survival in patients with glioblastoma multiforme of the cerebral hemisphere. Surg Neurol (1999) 52:371–9.10.1016/S0090-3019(99)00103-2
    1. Han SJ, Rutledge WC, Molinaro AM, Chang SM, Clarke JL, Prados MD, et al. The effect of timing of concurrent chemoradiation in patients with newly diagnosed glioblastoma. Neurosurgery (2015) 77:248–53.10.1227/NEU.0000000000000766
    1. Reardon DA, Gilbert MR, Wick W, Liau L. Immunotherapy for neuro-oncology: the critical rationale for combinatorial therapy. Neuro Oncol (2015) 17:vii32–40.10.1093/neuonc/nov178
    1. Neagu MR, Reardon DA. An update on the role of immunotherapy and vaccine strategies for primary brain tumors. Curr Treat Options Oncol (2015) 16:54.10.1007/s11864-015-0371-3
    1. Speiser DE, Ho PC, Verdeil G. Regulatory circuits of T cell function in cancer. Nat Rev Immunol (2016) 16:599–611.10.1038/nri.2016.80

Source: PubMed

3
S'abonner