We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Combating immunosuppression in glioma

    Eleanor A Vega

    Duke University School of Medicine, Department of Surgery, Division of Neurosurgery, 221 Sands Building, Durham, NC 27710, USA

    ,
    Michael W Graner

    Duke University School of Medicine, Department of Surgery, Division of Neurosurgery, 221 Sands Building, Durham, NC 27710, USA

    &
    John H Sampson

    † Author for correspondence

    Duke University School of Medicine, Department of Surgery, Division of Neurosurgery, 221 Sands Building, Durham, NC 27710, USA.

    Published Online:https://doi.org/10.2217/14796694.4.3.433

    Despite maximal therapy, malignant gliomas have a very poor prognosis. Patients with glioma express significant immune defects, including CD4 lymphopenia, increased fractions of regulatory T cells in peripheral blood and shifts in cytokine profiles from Th1 to Th2. Recent studies have focused on ways to combat immunosuppression in patients with glioma as well as in animal models for glioma. We concentrate on two specific ways to combat immunosuppression: inhibition of TGF-β signaling and modulation of regulatory T cells. TGF-β signaling can be interrupted by antisense oligonucleotide technology, TGF-β receptor I kinase inhibitors, soluble TGF-β receptors and antibodies against TGF-β. Regulatory T cells have been targeted with antibodies against T-cell markers, such as CD25, CTLA-4 and GITR. In addition, vaccination against Foxp3 has been explored. The results of these studies have been encouraging; combating immunosuppression may be one key to improving prognosis in malignant glioma.

    Papers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers.

    Bibliography

    • Stupp R, Mason WP, van den Bent MJ et al.: Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med.352,987–996 (2005).
    • De Vleeschouwer S, van Gool SW, Van Calenbergh F: Immunotherapy for malignant gliomas: emphasis on strategies of active specific immunotherapy using autologous dendritic cells. Childs Nerv. Syst.21,7–18 (2005).
    • Bechmann I, Galea I, Perry VH: What is the blood–brain barrier (not)? Trends Immunol.28,5–11 (2007).
    • Bechmann I, Priller J, Kovac A et al.: Immune surveillance of mouse brain perivascular spaces by blood-borne macrophages. Eur. J. Neurosci.14,1651–1658 (2001).
    • Poduslo JF, Ramakrishnan M, Holasek SS et al.: In vivo targeting of antibody fragments to the nervous system for Alzheimer’s disease immunotherapy and molecular imaging of amyloid plaques. J. Neurochem.102,420–433 (2007).
    • Engelhardt B, Ransohoff RM: The ins and outs of T-lymphocyte trafficking to the CNS: anatomical sites and molecular mechanisms. Trends Immunol.26,485–495 (2005).
    • Carson MJ, Doose JM, Melchior B, Schmid CD, Ploix CC: CNS immune privilege: hiding in plain sight. Immunol. Rev.213,48–65 (2006).
    • Perry VH: A revised view of the central nervous system microenvironment and major histocompatibility complex class II antigen presentation. J. Neuroimmunol.90,113–121 (1998).
    • Galea I, Bechmann I, Perry VH: What is immune privilege (not)? Trends Immunol.28,12–18 (2007).
    • 10  Fecci PE, Mitchell DA, Whitesides JF et al.: Increased regulatory T-cell fraction amidst a diminished CD4 compartment explains cellular immune defects in patients with malignant glioma. Cancer Res.66,3294–3302 (2006).• Despite decreased absolute counts of CD4+ T cells and Tregs in peripheral blood of patients with malignant glioma, Treg fraction of remaining CD4+ compartment is increased. Increased Treg fraction correlates to immune deficits associated with glioma in vitro; Treg removal reverses these immune defects.
    • 11  Dix AR, Brooks WH, Roszman TL, Morford LA: Immune defects observed in patients with primary malignant brain tumors. J. Neuroimmunol.100,216–232 (1999).
    • 12  Mahaley MS Jr, Brooks WH, Roszman TL, Bigner DD, Dudka L, Richardson S: Immunobiology of primary intracranial tumors. Part 1: studies of the cellular and humoral general immune competence of brain-tumor patients. J. Neurosurg.46,467–476 (1977).
    • 13  Young HF, Sakalas R, Kaplan AM: Inhibition of cell-mediated immunity in patients with brain tumors. Surg. Neurol.5,19–23 (1976).
    • 14  Brooks WH, Netsky MG, Normansell DE, Horwitz DA: Depressed cell-mediated immunity in patients with primary intracranial tumors. Characterization of a humoral immunosuppressive factor. J. Exp. Med.136,1631–1647 (1972).
    • 15  Elliott L, Brooks W, Roszman T: Role of interleukin-2 (IL-2) and IL-2 receptor expression in the proliferative defect observed in mitogen-stimulated lymphocytes from patients with gliomas. J. Natl Cancer Inst.78,919–922 (1987).
    • 16  Ashkenazi E, Deutsch M, Tirosh R, Weinreb A, Tsukerman A, Brodie C: A selective impairment of the IL-2 system in lymphocytes of patients with glioblastomas: increased level of soluble IL-2R and reduced protein tyrosine phosphorylation. Neuroimmunomodulation4,49–56 (1997).
    • 17  Elliott LH, Brooks WH, Roszman TL: Inability of mitogen-activated lymphocytes obtained from patients with malignant primary intracranial tumors to express high affinity interleukin 2 receptors. J. Clin. Invest.86,80–86 (1990).
    • 18  Brooks WH, Roszman TL, Rogers AS: Impairment of rosette-forming T lymphoctyes in patients with primary intracranial tumors. Cancer37,1869–1873 (1976).
    • 19  Young HF, Kaplan AM: Cellular immune deficiency in patients with glioblastoma. Surg. Forum27,476–478 (1976).
    • 20  Elliott L, Brooks W, Roszman T: Inhibition of anti-CD3 monoclonal antibody-induced T-cell proliferation by dexamethasone, isoproterenol, or prostaglandin E2 either alone or in combination. Cell Mol. Neurobiol.12,411–427 (1992).
    • 21  Roszman TL, Brooks WH: Immunobiology of primary intracranial tumours. III. Demonstration of a qualitative lymphocyte abnormality in patients with primary brain tumours. Clin. Exp. Immunol.39,395–402 (1980).
    • 22  Roszman TL, Brooks WH, Elliott LH: Immunobiology of primary intracranial tumors. VI. Suppressor cell function and lectin-binding lymphocyte subpopulations in patients with cerebral tumors. Cancer50,1273–1279 (1982).
    • 23  Huettner C, Czub S, Kerkau S, Roggendorf W, Tonn JC: Interleukin 10 is expressed in human gliomas in vivo and increases glioma cell proliferation and motility in vitro. Anticancer Res.17,3217–3224 (1997).
    • 24  Brenner AV, Linet MS, Fine HA et al.: History of allergies and autoimmune diseases and risk of brain tumors in adults. Int. J. Cancer99,252–259 (2002).
    • 25  Wiemels JL, Wiencke JK, Sison JD, Miike R, McMillan A, Wrensch M: History of allergies among adults with glioma and controls. Int. J. Cancer98,609–615 (2002).
    • 26  Wheeler CJ, Black KL, Liu G et al.: Thymic CD8+ T cell production strongly influences tumor antigen recognition and age-dependent glioma mortality. J. Immunol.171,4927–4933 (2003).
    • 27  Liau LM, Prins RM, Kiertscher SM 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.11,5515–5525 (2005).
    • 28  Akasaki Y, Liu G, Chung NH, Ehtesham M, Black KL, Yu JS: Induction of a CD4+ T regulatory type 1 response by cyclooxygenase-2-overexpressing glioma. J. Immunol.173,4352–4439 (2004).
    • 29  Fujita M, Zhu X, Sasaki K et al.: Inhibition of STAT3 promotes the efficacy of adoptive transfer therapy using type-1 CTLs by modulation of the immunological microenvironment in a murine intracranial glioma. J. Immunol.180,2089–2098 (2008).
    • 30  Parsa AT, Waldron JS, Panner A et al.: Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma. Nat. Med.13,84–88 (2007).
    • 31  Massague J: TGF-β signal transduction. Annu. Rev. Biochem.67,753–791 (1998).
    • 32  Platten M, Wick W, Weller M: Malignant glioma biology: role for TGF-β in growth, motility, angiogenesis, and immune escape. Microsc. Res. Tech.52,401–410 (2001).
    • 33  Hunter KE, Sporn MB, Davies AM: Transforming growth factor-βs inhibit mitogen-stimulated proliferation of astrocytes. Glia7,203–211 (1993).
    • 34  Rich JN, Zhang M, Datto MB, Bigner DD, Wang XF: Transforming growth factor-β-mediated p15(INK4B) induction and growth inhibition in astrocytes is SMAD3-dependent and a pathway prominently altered in human glioma cell lines. J. Biol. Chem.274,35053–35058 (1999).
    • 35  Vergeli M, Mazzanti B, Ballerini C, Gran B, Amaducci L, Massacesi L: Transforming growth factor-β 1 inhibits the proliferation of rat astrocytes induced by serum and growth factors. J. Neurosci. Res.40,127–133 (1995).
    • 36  Wesolowska A, Kwiatkowska A, Slomnicki L et al.: Microglia-derived TGF-β as an important regulator of glioblastoma invasion – an inhibition of TGF-β-dependent effects by shRNA against human TGF-β type II receptor. Oncogene27(7),918–930 (2007).
    • 37  Paulus W, Baur I, Huettner C et al.: Effects of transforming growth factor-β 1 on collagen synthesis, integrin expression, adhesion and invasion of glioma cells. J. Neuropathol. Exp. Neurol.54,236–244 (1995).
    • 38  Koochekpour S, Merzak A, Pilkington GJ: Vascular endothelial growth factor production is stimulated by gangliosides and TGF-β isoforms in human glioma cells in vitro. Cancer Lett.102,209–215 (1996).
    • 39  Yamagiwa S, Gray JD, Hashimoto S, Horwitz DA: A role for TGF-β in the generation and expansion of CD4+CD25+ regulatory T cells from human peripheral blood. J. Immunol.166,7282–7289 (2001).
    • 40  Fantini MC, Becker C, Monteleone G, Pallone F, Galle PR, Neurath MF: Cutting edge: TGF-β induces a regulatory phenotype in CD4+CD25- T cells through Foxp3 induction and down-regulation of Smad7. J. Immunol.172,5149–5153 (2004).
    • 41  de Visser KE, Kast WM: Effects of TGF-β on the immune system: implications for cancer immunotherapy. Leukemia13,1188–1199 (1999).
    • 42  Liu Y, Wang Q, Kleinschmidt-DeMasters BK, Franzusoff A, Ng KY, Lillehei KO: TGF-β2 inhibition augments the effect of tumor vaccine and improves the survival of animals with pre-established brain tumors. J. Neurooncol.81,149–162 (2007).
    • 43  Hau P, Jachimczak P, Schlingensiepen R et al.: Inhibition of TGF-β2 with AP 12009 in recurrent malignant gliomas: from preclinical to Phase I/II studies. Oligonucleotides17,201–212 (2007).• AP 12009 is an antisense oligodeoxynucleotide that binds to TGF-β2 mRNA and prevents its translation into a gene product. In Phase I/II clinical trials, intratumoral infusions of AP 12009 were shown to be safe and well-tolerated in patients with recurrent malignant glioma. Several patients experienced prolonged survival.
    • 44  Uhl M, Aulwurm S, Wischhusen J et al.: SD-208, a novel transforming growth factor β receptor I kinase inhibitor, inhibits growth and invasiveness and enhances immunogenicity of murine and human glioma cells in vitro and in vivo. Cancer Res.64,7954–7961 (2004).• In a murine model for glioma, the TGF-β receptor I kinase inhibitor, SX-007, increased median survival time and reversed some of the TGF-β-induced immune defects associated with glioma.
    • 45  Tran TT, Uhl M, Ma JY et al.: Inhibiting TGF-β signaling restores immune surveillance in the SMA-560 glioma model. Neuro Oncol.9,259–270 (2007).
    • 46  Wojtowicz-Praga S: Reversal of tumor-induced immunosuppression by TGF-β inhibitors. Invest. New Drugs21,21–32 (2003).
    • 47  Dong M, How T, Kirkbride KC et al.: The type III TGF-β receptor suppresses breast cancer progression. J. Clin. Invest.117,206–217 (2007).
    • 48  Hempel N, How T, Dong M, Murphy SK, Fields TA, Blobe GC: Loss of β-glycan expression in ovarian cancer: role in motility and invasion. Cancer Res.67,5231–5238 (2007).
    • 49  Sharifi N, Hurt EM, Kawasaki BT, Farrar WL: TGFBR3 loss and consequences in prostate cancer. Prostate67,301–311 (2007).
    • 50  Biswas S, Guix M, Rinehart C et al.: Inhibition of TGF-β with neutralizing antibodies prevents radiation-induced acceleration of metastatic cancer progression. J. Clin. Invest.117,1305–1313 (2007).
    • 51  Nam JS, Suchar AM, Kang MJ et al.: Bone sialoprotein mediates the tumor cell-targeted prometastatic activity of transforming growth factor β in a mouse model of breast cancer. Cancer Res.66,6327–6335 (2006).
    • 52  Liu VC, Wong LY, Jang T et al.: Tumor evasion of the immune system by converting CD4+CD25- T cells into CD4+CD25+ T regulatory cells: role of tumor-derived TGF-β. J. Immunol.178,2883–2892 (2007).
    • 53  Nomura T, Sakaguchi S: Naturally arising CD25+CD4+regulatory T cells in tumor immunity. Curr. Top. Microbiol. Immunol.293,287–302 (2005).
    • 54  Randolph DA, Fathman CG: Cd4+Cd25+ regulatory T cells and their therapeutic potential. Annu. Rev. Med.57,381–402 (2006).
    • 55  Baecher-Allan C, Anderson DE: Regulatory cells and human cancer. Semin. Cancer Biol.16,98–105 (2006).
    • 56  Thornton, AM, Shevach EM: CD4+CD25+ immunoregulatory T cells suppress polyclonal T cell activation in vitro by inhibiting interleukin 2 production. J. Exp. Med.188,287–296 (1998).
    • 57  Piccirillo CA, Shevach EM: Cutting edge: control of CD8+ T cell activation by CD4+CD25+ immunoregulatory cells. J. Immunol.167,1137–1140 (2001).
    • 58  Dieckmann D, Bruett CH, Ploettner H, Lutz MB, Schuler G: Human CD4+CD25+ regulatory, contact-dependent T cells induce interleukin 10-producing, contact-independent type 1-like regulatory T cells [corrected]. J. Exp. Med.196,247–253 (2002).
    • 59  Liyanage UK, Moore TT, Joo HG et al.: Prevalence of regulatory T cells is increased in peripheral blood and tumor microenvironment of patients with pancreas or breast adenocarcinoma. J. Immunol.169,2756–2761 (2002).
    • 60  Ichihara F, Kono K, Takahashi A, Kawaida H, Sugai H, Fujii H: Increased populations of regulatory T cells in peripheral blood and tumor-infiltrating lymphocytes in patients with gastric and esophageal cancers. Clin. Cancer Res.9,4404–4408 (2003).
    • 61  Rosenberg SA, Sportes C, Ahmadzadeh M et al.: IL-7 administration to humans leads to expansion of CD8+ and CD4+ cells but a relative decrease of CD4+ T-regulatory cells. J. Immunother. (1997) 29,313–319 (2006).
    • 62  Fecci PE, Sweeney AE, Grossi PM et al.: Systemic anti-CD25 monoclonal antibody administration safely enhances immunity in murine glioma without eliminating regulatory T cells. Clin. Cancer Res.12,4294–4305 (2006).
    • 63  Grauer OM, Nierkens S, Bennink E et al.: CD4+FoxP3+ regulatory T cells gradually accumulate in gliomas during tumor growth and efficiently suppress antiglioma immune responses in vivo. Int. J. Cancer121,95–105 (2007).• Glioma-bearing mice show a decrease in CD4+ T cells and Treg absolute number in peripheral blood, while Tregfraction is increased; the reverse is seen in bone marrow. In a murine model for glioma, systemic administration of anti-CD25 mAb inhibited the suppressive action of Tregs, without completely eliminating the Treg fraction. Combining anti-CD25 mAb with tumor RNA-pulsed dendritic cell vaccination led to tumor rejection in 100% of mice.
    • 64  Powell DJ Jr, Felipe-Silva A, Merino MJ et al.: Administration of a CD25-directed immunotoxin, LMB-2, to patients with metastatic melanoma induces a selective partial reduction in regulatory T cells in vivo. J. Immunol.179,4919–4928 (2007).
    • 65  Ochiai H, Archer GE, Herndon JE 2nd et al.: EGFRvIII-targeted immunotoxin induces antitumor immunity that is inhibited in the absence of CD4+ and CD8+ T cells. Cancer Immunol. Immunother.57(1),115–121 (2007).
    • 66  Litzinger MT, Fernando R, Curiel TJ, Grosenbach DW, Schlom J, Palena C: The IL-2 immunotoxin denileukin diftitox reduces regulatory T cells and enhances vaccine-mediated T-cell immunity. Blood110(9),3192–3201 (2007).
    • 67  Mahnke K, Schonfeld K, Fondel S et al.: Depletion of CD4+CD25+ human regulatory T cells in vivo: kinetics of Treg depletion and alterations in immune functions in vivo and in vitro. Int. J. Cancer120,2723–2733 (2007).
    • 68  Attia P, Maker AV, Haworth LR, Rogers-Freezer L, Rosenberg SA: Inability of a fusion protein of IL-2 and diphtheria toxin (Denileukin Diftitox, DAB389IL-2, ONTAK) to eliminate regulatory T lymphocytes in patients with melanoma. J. Immunother.28,582–592 (2005).
    • 69  Cranmer LD, Hersh E: The role of the CTLA4 blockade in the treatment of malignant melanoma. Cancer Invest.25,613–631 (2007).
    • 70  Fecci PE, Ochiai H, Mitchell DA et al.: Systemic CTLA-4 blockade ameliorates glioma-induced changes to the CD4+ T cell compartment without affecting regulatory T-cell function. Clin. Cancer Res.13,2158–2167 (2007).• Systemic administration of monoclonal antibody to CTLA-4 led to prolonged survival in a murine model for glioma. CTLA-4 blockade also re-established normal CD4 counts and proliferative activity and prevented increase in Treg fraction; benefits were due to effect on CD4+CD25- T cells only, and not on Tregs.
    • 71  Phan GQ, Yang JC, Sherry RM et al.: Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma. Proc. Natl Acad. Sci. USA100,8372–8377 (2003).
    • 72  Attia P, Phan GQ, Maker AV et al.: Autoimmunity correlates with tumor regression in patients with metastatic melanoma treated with anti-cytotoxic T-lymphocyte antigen-4. J. Clin. Oncol.23,6043–6053 (2005).
    • 73  Maker AV, Phan GQ, Attia P et al.: Tumor regression and autoimmunity in patients treated with cytotoxic T lymphocyte-associated antigen 4 blockade and interleukin 2: a Phase I/II study. Ann. Surg. Oncol.12,1005–1016 (2005).
    • 74  Nocentini G, Ronchetti S, Cuzzocrea S, Riccardi C: GITR/GITRL: more than an effector T cell co-stimulatory system. Eur. J. Immunol.37,1165–1169 (2007).
    • 75  Ko K, Yamazaki S, Nakamura K et al.: Treatment of advanced tumors with agonistic anti-GITR mAb and its effects on tumor-infiltrating Foxp3+CD25+CD4+ regulatory T cells. J. Exp. Med.202,885–891 (2005).
    • 76  Tuyaerts S, van Meirvenne S, Bonehill A et al.: Expression of human GITRL on myeloid dendritic cells enhances their immunostimulatory function but does not abrogate the suppressive effect of CD4+CD25+ regulatory T cells. J. Leukoc. Biol.82,93–105 (2007).
    • 77  Nair S, Boczkowski D, Fassnacht M, Pisetsky D, Gilboa E: Vaccination against the forkhead family transcription factor Foxp3 enhances tumor immunity. Cancer Res.67,371–380 (2007).• Vaccination against foxp3 in a murine model of glioma led to a Foxp3-specific CTL response and a reduction in number of Tregsintratumorally, but not in the periphery, whereas administration of anti-CD25 antibody caused Tregdepletion in both locations.
    • 78  Heimberger AB, Sun W, Hussain SF et al.: Immunological responses in a patient with glioblastoma multiforme treated with sequential courses of temozolomide and immunotherapy: case study. Neuro. Oncol.10,98–103 (2008).
    • 79  Comes A, Rosso O, Orengo AM et al.: CD25+ regulatory T cell depletion augments immunotherapy of micrometastases by an IL-21-secreting cellular vaccine. J. Immunol.176,1750–1758 (2006).
    • 101  Description of the Safety and Efficacy Study of GC1008 to Treat Renal Cell Carcinoma or Malignant Melanoma from the ClinicalTrials.gov website http://clinicaltrials.gov/ct/show/NCT00356460?order=2)