1. Introduction
Glioblastoma multiforme (GBM) is the most common and most aggressive adult brain tumor with a patient median survival of 15 months from the time of diagnosis, and less than 20 weeks for patients with recurrent tumors.Current standard of care consists of multi-modality therapy including image-guided tumor resection, fractionated radiotherapy, and chemotherapy.This aggressive therapy is non-specific and highly toxic, leaving collateral damage to surrounding normal brain and systemic tissue, and is often debilitating to patients.Thus, there is a dire need for a more effective therapy that more specifically targets tumor cells while minimizing damage to surrounding eloquent cerebral cortex.Immunotherapy is based on the premise that the inherent sensitivity and specificity of immunologic reactivity could deliver tumor cell-specific therapy.Cellular immunotherapy aims to utilize the patient’s own immune cells that are harvested, expanded
During early efforts in immunotherapy, tumor specific antigens were unknown and it was unclear whether tumor antigens could be recognized and targeted by the immune system.The identification of tumor antigens began with those expressed in malignant melanoma, and soon there was an explosion in the development of antigen specific immunologic treatments against solid tumors. In the past several years, pre-clinical models of cancer have reliably demonstrated that the immune system is capable of targeting tumor antigens and eradicating malignancies. It has also been demonstrated clinically that the human immune system is capable of recognizing antigens within malignant tumor cells with precision, and current immunotherapy research aims to induce potent antitumor immune responses to prolong patient survival.It was initially unclear if a potent immune response was inducible against brain tumors because of the immunoprivileged nature of the nervous system, but studies have demonstrated that immune effector cells can infiltrate the central nervous system (CNS) and induce efficient immune responses against intracranial tumors.
Current research in cellular immunotherapy against cancer is directed at eliciting a specific immune response against tumor antigens using active immunization with cellular vaccines or adoptive transfer of
2. Immune privilege
Cellular immune responses must afford protection without causing collateral damage to normal tissue.This is particularly important in the brain where passive and active mechanisms maintain a state of immunological privilege that limits the magnitude of the immune response.It has been demonstrated that immune responses in the CNS can be induced, the magnitude of this response is strictly regulated by the presence of the blood-brain barrier.Cerebral interstitial fluid (CIF) is secreted at the blood-brain barrier and flows within the spaces of the brain parenchyma.Cerebrospinal fluid (CSF) is formed by the choroid plexus within the ventricles and subarachnoid membrane, then flows through the ventricles to the basal cisterns, then through the subarachnoid space [1-3].Antigens within the CNS enter the lymph nodes via the CSF which drains into the Virchow-Robbin spaces to the deep cervical lymphatic’s via perivascular sheaths and through the subnasal mucosa [2, 4, 5].The flow of CSF exits the subarachnoid space through the arachnoid granulations and through drainage along the olfactory nerve across the cribriform plate into blood circulation and cervical lymph nodes [4, 6, 7]. Antigens draining to cervical lymph nodes encounter cognate B cells and can also be processed and presented to T cells [4, 6, 7].
Immune activation occurs with a distinct hierarchy in terms of the types of responses induced [6].Antigens that drain into the periphery via the cervical lymph nodes induce a response characteristic of a strong antibody response and the priming of cytotoxic T cell responses, but an absence of delayed-type hypersensitivity (DTH) responses with a skewing towards a Th2 phenotype [1, 2, 6, 8].Strong humoral responses are induced in response to antigenic challenge.T cells are not endogenously found in the brain, but T cells and antibodies [9] have access to antigens in the brain, indicating that the blood-brain barrier does not entirely prohibit immune responses.Activated T cells “patrol” the CNS and return to systemic circulation, exiting through the cribriform plate, through the nasal mucosa, and then the cervical lymph nodes [1, 10].Some studies suggest that T cells that encounter their cognate antigen are retained within the CNS [11], but do not proliferate and undergo apoptosis [12].Alternatively, other studies have demonstrated that T cells encountering cognate antigen proliferate and differentiate into tumor-specific T cells, with enhanced effector function [1].
Professional antigen presenting cells (APC) such as DCs have not been described in the CNS.Microglia are the resident antigen presenting cells in the CNS, but DCs are present in the choroid plexus and meninges [10, 11, 13, 14].Immunologic responses in the CNS require complex interactions between resident immune cells such as microglia and astrocytes, and peripheral macrophages, lymphocytes, and DCs [14-18]. Microglia constitutively express MHC class II antigens and T cell co-stimulatory molecules.Microglia are bone marrow derived cells that are capable of presenting antigen to T helper cells
3. Glioma Immunology
In the past decade, tumor-associated antigens that are recognizable by cytotoxic T lymphocytes (CTL) have been identified and have the been the basis of cancer immunotherapy.In cancer patients, tumor-specific endogenous immunity can be elicited when tumor antigens are overexpressed, however the immune response is incapable of preventing tumor growth. The immunosuppressive tumor microenvironment, the low avidity of the T cells for tumors, and the low grade immune response are all contributing factors to the inhibition of the endogenous antitumor response. Glioma cells secrete immunosuppressive cytokines including transforming growth factor beta (TGF-) and vascular endothelial growth factor (VEGF) [20-22] that contribute to tumor immune evasion.In addition, the increased frequency of T regulatory cells in tumor bearing patients plays a critical role in tumor tolerance [23-25].
Cancer vaccines are designed to augment patient immunity by boosting low-level immunity and stimulating the proliferation of higher-avidity T cells. Clinical studies have reported that immunotherapy by systemic administration of antigen-specific DCs and peptide antigens is capable of inducing an antitumor response against malignancies, including CNS malignancies [26-30].
In 1991, van der Bruggen
4. Immunosuppression in GBM
Patients with brain malignancies have impaired B and T cell immune function in part due to tumor secreted factors, but greatly due to depressed cellular immunity and increased levels of T regulatory cells [25, 44].T regulatory cell frequency is increased CD4+ T cell subset in lymphopenic patients bearing malignant gliomas [25, 45].Peripheral blood lymphocytes from glioma patients proliferate poorly in response to T cell mitogens, anti CD3, and T and B cell dependent mitogens [46-48].The total T cell compartment has limited capabilities to respond to mitogen stimulation [46, 47, 49, 50].
4.1. Immunosuppressive cytokines
Two immunosuppressive cytokines secreted by gliomas are TGF- and VEGF. TGF- has been isolated from malignant glioma cell supernatants, and the gene encoding for TGF-2 was cloned from a glioma cell line [51]. TGF- suppresses the generation of cytotoxic T lymphocytes from PBLs and tumor-infiltrating lymphocytes by inhibiting IL-2 receptor expression on T cells, reducing IL-1 and IL-2, and depressing natural killer cell activation. TGF- also inhibits the differentiation of cytotoxic T lymphocytes, reduces IFN production, and downregulates MHC class II-dependent antigen expression [52, 53].In an
VEGF is produced by most solid tumor cells and plays an important role in tumor immunosuppression by inhibiting the maturation of bone marrow derived DCs [55, 56] by inhibiting NF-KB signaling in hematopoietic progenitor cells.In the context of DC vaccination in tumor bearing mice, inhibition of VEGF production with a blocking anti-VEGF monoclonal antibody enhanced antitumor efficacy [57], demonstrating that attenuating VEGF-mediated immunosuppression is vital to proper function of immunotherapy.VEGF and TGF- production by tumors contribute to tumor vascularization and immune evasion, contributing to the systemic immunosuppression found in glioma patients.Monoclonal antibodies against VEGF are used therapeutically (bevacizumab) and have been shown to be efficacious against malignant gliomas [58-60].Preclinical studies conducted in xenogeneic systems with human brain tumor bearing immunodeficient mice have demonstrated that inhibition of VEGF is efficient in prohibiting angiogenesis, leading to subsequent growth suppression of tumors [61]
4.2. T regs
The CD4+FOXP3+CD25+ T regulatory cell subset normally comprises of 5-10% of the total CD4+ compartment [62-64].T regulatory cells inhibit T cell cytokine secretion while inhibiting endogenous or induced immune responses [65, 66].T regulatory cells play a significant role in hindering immunity to normal and tumor antigens [67, 68], and represent an increased frequency of CD4+ cells in the peripheral blood of GBM patients [44].Targeting T regulatory cell activity to counter their immunosuppressive effects enhances antitumor immunity in murine and human hosts.Fecci
5. Immunotherapy
5.1. Antibody-based immunotherapy
Therapeutic use of antibodies aims to alter patient immunity by delivering monoclonal antibodies (mAb) that are targeted against TSAs or TAAs. Antitumor antibodies have been used as either naked antibodies or as vehicles to deliver radioisotopes or toxins to tumors.It is imperative that the mAb can recognize and bind to tumor tissue with high specificity and affinity, without accumulation in normal tissue.Antibody based immunotherapy has been successful for lymphomas (rituximab) and breast cancer (trastuzumab).Bevacizumab, a monoclonal antibody against the angiogenic regulator, vascular endothelial growth factor (VEGF), was approved by the FDA for the treatment of recurrent glioblastoma in 2009 [69].Blocking VEGF is effective in normalizing abnormal tumor vasculature and increasing tumor response to radiation and chemotherapy [70].
EGFRvIII is currently the only TSA found on malignant glioma cells, but is absent from normal brain tissue.EGFRvIII consists of an in-frame deletion of exons 2-7 from the extracellular domain of the EGFR that splits a codon and produces a novel glycine at the fusion junction [71, 72].The new glycine inserted at the fusion junction of normally distant parts of the extracellular domain results in a tumor-specific epitope not found in any normal tissue.This tumor-specific mutation encodes a constitutively active tyrosine kinase that enhances tumorigenicity [73-75] and migration of tumor cells that confers radiation and chemotherapy resistance [76-78].The EGFRvIII mutation is expressed on the plasma membrane of up to 100% of glioma cells and is frequently found in GBM patients [79, 80].Through the use of reverse transcriptase-polymerase chain reaction (RT-PCR) and fluorescent in situ hybridization (FISH) studies have detected the EGFRvIII mutation on 6-21% of grade III/IV gliomas that have amplified EGFR [80-82]. In addition, analysis using FACS found EGFRvIII expression in 50% of GBM samples [83].The expression of this mutation confers a negative prognosis for GBM patients.The tumor-specific clonal expression of EGFRvIII on GBMs and its absence from normal tissues make EGFRvIII an ideal target for anti-tumor immunotherapy.
In pre-clinical systems, EGFRvIII expressing cell lines or PEPvIII, an EGFRvIII-specific 14-amino acid peptide, has been used for the generation of EGFRvIII-specific antibodies [79, 84-86], induction of cellular immune responses, or derivation of targeted toxins [87, 88].Both murine and human chimeric EGFRvIII antibodies have been cloned for use in diagnostic immunohistochemistry and FACS [79].Monoclonal antibodies binding EGFRvIII are rapidly internalized and have been successfully used
It has long been established that EGFR and its downstream signaling pathway plays a role in oncogenesis and tumor progression in malignant brain tumors. Thus arose efforts to block the EGFR pathway with the aim of inhibiting tumor cell proliferation with anti-EGFR monoclonal antibodies developed for clinical use.Faillot
EGFRvIII has also been shown to be immunogenic in humans [94].While anti-EGFRvIII antibodies have not been identified in normal volunteers, patients with malignant gliomas develop EGFRvIII specific antibodies.Weak CTL epitopes restricted by MHC class I and class II have been identified and are sufficient to induce EGFRvIII-specific lymphocyte proliferation and cytokine production.Phase I/II clinical trials targeting this mutation demonstrated that vaccines targeting EGFRvIII are capable of inducing antitumor immunity.In a phase II multicenter trial between Duke University Medical Center and M.D. Anderson Cancer Center (FDA BB-IND-9944), 18 patients with EGFRvIII expressing primary GBMs were treated with an EGFRvIII peptide vaccine called PEPvIII, which is a 13- amino-acid peptide with an additional terminal cysteine that spans the entire
5.2. Radiolabeled antibodies
Unlabelled antibodies can be used as delivery vehicles to administer effector molecules such as toxins or radiation directly to tumors.The specificity of tumor associated antigens guide molecules to targets using the specificity of antibodies.The most common effectors conjugated to antibodies are radionucleotides.Despite the expression of EGFRvIII, tenascin has been the most widely evaluated antigenic target.Tenascin is an extracellular matrix protein that is highly expressed in gliomas [97] and its expression increases with tumor progression and is a logical target of trials using radioimmunotherapy.Conjugating antibodies with radioisotopes has been a focus in clinical studies.
The antibody 81C6 is a radiolabeled antibody used in a number of clinical studies [98-102].81C6 reacts with an alternatively spliced segment of tenascin at the fibronectin type III domain.Its tumor reactivity and specificity to gliomas is superior to other anti-glioma mAbs and has been proven to be clinically safe.In a safety study at Duke University, antitenascin 81C6 labeled with 131-I was administered into the surgical resection cavity of 21 newly diagnosed GBM patients to achieve a 44-Gy boost specifically to the 2-cm margin of the resection cavity [103].In 17 patients, 131-I was administered prior to external beam radiotherapy (XRT), and 3 patients 131-I was administered after XRT.Conventional XRT and chemotherapy was then administered.One patient opted not to receive XRT or chemotherapy.Twenty out of twenty-one total patients enrolled received the targeted 44-Gy boost and at a median follow-up of 151 weeks, medial overall survival times for all patients was 96.6 weeks [103]. This study demonstrated that this radioimmunotherapy was well tolerated with encouraging survival in patients with malignant gliomas.Other studies have demonstrated that 81C6 increased survival in patients with leptomeningeal neoplasm as well as recurrent and newly diagnosed gliomas [98, 99, 101, 102].In a study conducted conducted at Duke University [102], 33 patients with previously untreated malignant glioma (GBM, n=27; anaplastic astrocytoma, n=4; anaplastic oligodendroglioma, n=2) were given 81C6 into the surgical resection cavity followed by conventional XRT and chemotherapy.The observed median survival for all patients was 86.7 weeks, and 79.4 weeks for GBM patients.The median survival of patients treated with 131-I in this study exceeded that of historical controls treated with conventional therapy.
211At is an alpha-emitting radionucleotide, and also emits K X-ray of sufficient energy to allow both -counting of tissue samples and external imaging [104]. This -emiting nucleotide is more advantageous to gliomas than other isotopes.For example, since damage to normal tissue in the brain is most detrimental to the patient’s cognitive function, specificity of isotope delivery is essential.The range of 211At particles is only up to 2 mm, thus toxicity is confined to the peritumoral area, minimizing collateral damage to normal tissue.211At -particles have a linear energy transfer that is ideal for maximizing biologic efficacy.The distance between ionizing events is approximately the distance between DNA strands, thus increasing the likelihood of inducing irreparable DNA breaks, thereby increasing cytotoxicity [104]. In a phase I safety study, 18 patients with histologic diagnosis of recurrent supratentorial primary malignant brain tumors were treated with 211At-labeled anti-tenascin mAb administered into the surgical resection cavity and treated with salvage chemotherapy [105].No toxicities of grade 3 or higher were observed.The median survival in patients with recurrent GBM was 54 weeks, patients with anaplastic astrocytoma or oligodendroglioma had a median survival of 52 and 116 weeks respectively. Local administration of 211At-81C6 is safe, feasible, and may potentially provide a survival benefit in recurrent malignant brain tumor patients.
5.3. Dendritic cells and tumor immunotherapy
DCs induce, regulate, and maintain T cell immunity and are essential for the foundation of immunotherapy [106, 107].DCs take-up and process antigens, thus playing a critical role in T cell priming and regulation of the immune response. DCs are equipped with antigen-processing machinery (APM) essential for uptake and processing of tumor-derived antigens so that tumor-derived epitopes can be cross-presented to T cells [20]. Immature (non-activated) DCs present self-antigens to T cells, inducing a tolerizing immune response by activating T regulatory cells [108].Immature DCs do not have the ability to stimulate naïve or antigen-specific T memory cells [109, 110]. Immature DCs can take-up antigens via receptor- or nonreceptor-mediated mechanisms.Upon internalization, tumor antigens are processed and split into peptides in the cytosol or endocytic vesicles, then expressed on the cell surface in association with MHC molecules [20, 111].
Activated mature antigen-loaded DCs are responsible for antigen-specific immune responses that lead to T cell activation and proliferation into T helper and effector cells [111].The two major DC subsets are the classical DCs (myeloid DCs) and plasmacytiod DCs.Plasmacytoid DCs are responsible for the antiviral immune response, producing high amounts of type I IFN/ in response to viruses [112].Classical DCs are further categorized in subsets displaying different phenotypes and functions.The skin contains Langerhans cell (LC) found in human epidermis, and the dermal layer contains two subsets, CD1a+ DCs and CD14+ DCs [113, 114]. CD14+ DCs are geared toward mounting humoral immunity.LCs prime high avidity antigen-specific CD8+ T lymphocytes [115].
A number of phase I safety and feasibility clinical studies have evaluated the use of antigen-loaded DC vaccination for the treatment of malignant glioma [26, 27, 119-121].Yu
EGFRvIII is an evident target for tumor-targeted immunotherapy since it is the only tumor-specific antigen in gliomas.Duke University Medical Center conducted a phase I clinical trial whereby 16 glioma patients received intradermal immunizations with autologous DCs pulsed with PEPvIII, a keyhole limpet hemocyanin (KLH) conjugate of a peptide spanning the mutated region of EGFRvIII.The logic follows that DCs injected intradermally will migrate to lymph nodes, subsequently presenting antigen to T lymphocytes [123, 124].The patients in this study were adults with malignant gliomas who underwentresection and radiotherapy. Patients underwent leukapheresis to collect autologous peripheral blood mononuclear cells from which to generate DCs
In a phase I trial, 12 GBM patients were given DCs pulsed with peptides eluted from the surface of resected autologous tumor in three bi-weekly intradermal injections [119].In addition to demonstrating no adverse events occurring after DC vaccinations, the study demonstrated increased systemic and intracranial immunologic responses against autologous tumor in 50% of treated patients with a median survival of 23.4 months[119].
De Vleeschouwer
Wheeler
Prins
5.4. RNA-pulsed DCs
Vaccine treatments dependent on large amounts of autologous tumor tissue can be limited in patients with brain tumors because of small amounts of material available after resection.Small amounts of tumor tissue is also a limitation to tumor-lysate based DC therapy because It has been argued that continuous boosting is required to maintain antitumor protection [130, 131]. The use of tumor antigen RNA-pulsed DCs demonstrably stimulates potent antitumor immunity in both murine and human cells [132, 133].Both murine and human tumor-derived RNA can be isolated and amplified without loss of function, thus an RNA based platform will not be limited by the availability of tumor tissue [132].RNA transfection has also been demonstrated to be a superior method for antigen-loading of DCs [134-136], in addition, RNA-loaded DCs have been found to be better stimulators of antigen-specific T cells than other methods of loading DCs [135].In an
In early studies with prostate cancer, DCs transfected with prostate-specific antigen RNA and were capable of inducing cytotoxic T lymphocyte responses specifically against prostate-specific antigens, but not kallikrein antigens, a protein that shares homology with prostate-specific antigens.This demonstrates the specificity of the elicited immune response [133].RNA-pulsed DC responses are not restricted to single MHC haplotype, nor a specific T cell subtype, enabling activation of both cytotoxic T lymphocytes and T helper cells [137-139].
In a phase I clinical study by Caruso
Preclinical murine models of tumor challenge have demonstrated that DCs pulsed with unselected tumor-derived antigens induce potent protective immune responses without toxicity due to autoimmunity [142-145]; however in studies modeling large solid tumors, much stronger immune responses were required for protection [146, 147]. When such responses were generated against tumor-associated antigens not exclusive to tumors, severe autoimmunity was observed in some but not all mice [147].This platform is capable of engendering a range of immune responses, and further studies are essential to find the balance between antitumor efficacy and prevention of toxicity.
Given the immense potential for the clinical use of DC-based tumor-specific immunotherapy, studies to examine strategies of maximizing DC potential are necessary.In the past decade, the ability of DC-based strategies to induce effective T-cell responses against malignant astrocytomas has been demonstrated using human DCs.DCs generated from tumor-bearing patients were fused with autologous tumor cells or pulsed with total tumor RNA or tumor lysate.Their respective abilities to generate a tumor-specific T cell proliferation and cytotoxic response
5.5. Adoptive cell transfer
Adoptive transfer involves the transfusion of cells that were manipulated
In 1992, Riddell
In 1984, Steinbok
5.6. LAK cells
Lymphokine-activated killer cells (LAK) are
In another clinical trial, 40 GBM patients received 2.0 ± 1.0 x 109 autologous LAK cells into their post-surgical cavity.The median interval from time of diagnosis to receiving LAK cell treatment was 10.9 months.The median survival from initial diagnosis for 31 GBM patients was 17.5 months [168].Although this trial did not have clear survival benefits, it demonstrated the safety and feasibility of adoptive transfer of
5.7. TILS and tumor-draining-lymph node T cells
In attempts to increase T cell specificity of adoptively transferred cells, Kitahara
Another means of isolating tumor-specific lymphocytes is to isolate lymphocytes directly from the tumor. Autologous tumor infiltrating lymphocytes (TILS) were first demonstrated to mediate tumor regression in melanoma in 1988 [163].In this early study, the response rate was 33%.Further studies in host preconditioning substantially increased the antitumor efficacy of TILS in melanoma [169], with clinical responses in up to 50% of patients.
The recovered TILS are found in the tumor by the time surgical resection occurs.These cells are already ‘primed’ against the tumor and thus have tumor-specific activation.In clinical studies, TILS were recovered from tumors and re-administered into the tumor post-surgical cavity in addition to IL-2 to enhance T cell proliferation.This was most studied in melanoma patients, but in a study by Quattrocchi
In another trial, Kruse
The adoptive transfer of
Incredible advances in adoptive immunotherapy have been made in metastatic melanoma to maximize the clinical benefits of adoptive transfer methods by optimizing host conditioning, genetic manipulation of T cells, and optimizing
6. Host conditioning and homeostatic proliferation
Lymphodepletion is well known to significantly enhance the antitumor efficacy of adoptive cell transfer and DC vaccination strategies in tumor bearing hosts.Lymphodepletion removes inhibitory T regulatory cells, decreases competition for homeostatic cytokines between host and transferred cells, and induces homeostatic proliferation of the few remaining host T lymphocytes. Homeostatic proliferation is a rapid expansion of T cells with the purpose of recovering normal lymphocyte counts [175].An increase in serum levels of IL-7 and IL-15 help induce rapid proliferation of T cells with a lower activation threshold [175, 176] and differentiate into effector memory T cells that respond to antigen [45].Lymphocytes must encounter cognate antigens and compete for these cytokines.Following this logic, B and T cells that are antigen-specific such as those provided as vaccines or as adoptively transferred antigen-specific T lymphocytes, have a competitive advantage over depleted host lymphocytes [177, 178].Antigen-specific lymphocytes disproportionately expand to become over-represented in the host circulation both in murine models and human patients [177-179], therefore enhancing antitumor immunity [177, 178, 180].
In preclinical and clinical studies of adoptive immunotherapy in metastatic melanoma, lymphodepletion enhanced the expansion of adoptively transferred tumor-specific T cells and resulted in increased clinical responses with a greater than 50% objective clinical response [174, 181-185].Adoptively transferred cells undergo dramatic expansion and can constitute up to 90% of host T cell repertoire and persist for months [174].These studies by Dudley and Rosenberg demonstrate a correlation between clinical regression of systemic disease, the frequency of tumor-specific T cells in peripheral blood, and the persistence of transferred cells
7. Conclusion
Cellular immunotherapy is a highly specific therapy that is directed at eliciting an immune response against tumor antigens using passive or active immunization with cellular vaccines or adoptive transfer of ex vivo activated lymphocytes.Preclinical studies have demonstrated the clear antitumor efficacy of these therapeutic modalities.The breadth of clinical studies conducted demonstrates a lack of adverse toxicity related to immunotherapies.The curative potential of cellular immunotherapy has been successful in other solid and hematological malignancies and is currently in the early stages of use in CNS malignancies.
References
- 1.
Heimberger, A.B. and J.H. Sampson, Immunotherapy coming of age: what will it take to make it standard of care for glioblastoma? Neuro Oncol, 2011 3 13 - 2.
Harling-Berg, C., et al., Role of cervical lymph nodes in the systemic humoral immune response to human serum albumin microinfused into rat cerebrospinal fluid. J Neuroimmunol,1989 185 193 - 3.
Cserr, H.F., et al., Afferent and efferent arms of the humoral immune response to CSF-administered albumins in a rat model with normal blood-brain barrier permeability. J Neuroimmunol,1992 195 202 - 4.
Cserr, H.F., C.J. Harling-Berg, and P.M. Knopf, Drainage of brain extracellular fluid into blood and deep cervical lymph and its immunological significance. Brain Pathol,1992 269 276 - 5.
Brain Pathol,Weller R. O. Engelhardt B. Phillips M. J. Lymphocyte targeting. of the. central nervous. system a. review of. afferent efferent C. N. S-immune pathways. 1996 275 288 - 6.
Harling-Berg, C.J., et al., Hierarchy of immune responses to antigen in the normal brain. Curr Top Microbiol Immunol,2002 1 22 - 7.
Fabry, Z., C.S. Raine, and M.N. Hart, Nervous tissue as an immune compartment: the dialect of the immune response in the CNS. Immunol Today,1994 218 224 - 8.
Mosmann, T.R. and R.L. Coffman, TH1 and TH2 cells: different patterns of lymphokine secretion lead to different functional properties. Annu Rev Immunol,1989 145 173 - 9.
Bullard, D.E. and D.D. Bigner, Applications of monoclonal antibodies in the diagnosis and treatment of primary brain tumors. J Neurosurg,1985 2 16 - 10.
J., et al., J Leukoc Biol,Goldmann J. et al. cells T. traffic from. brain to. cervical lymph. nodes via. the cribroid. plate the nasal. mucosa 2006 797 801 - 11.
J Immunol,Masson F. et al. Brain microenvironment. promotes the. final functional. maturation of. tumor-specific effector. C. D. cells T. 2007 845 853 - 12.
Immunol Today,Aloisi F. Ria F. Adorini L. Regulation of. T-cell responses. by C. N. S. antigen-presenting cells. different roles. for microglia. astrocytes 2000 141 147 - 13.
Serot, J.M., et al., Ultrastructural and immunohistological evidence for dendritic-like cells within human choroid plexus epithelium. Neuroreport,1997 1995 1998 - 14.
Brain Res Brain Res Rev,Gehrmann J. Matsumoto Y. Kreutzberg G. W. Microglia intrinsic. immuneffector cell. of the. brain 1995 269 287 - 15.
J Exp Med,Ford A. L. et al. Microglia induce. C. D. T. lymphocyte final. effector function. death 1996 1737 1745 - 16.
J.D., et al., J Immunol,Sedgwick J. D. et al. Central nervous. system microglial. cell activation. proliferation follows. direct interaction. with tissue-infiltrating. T. cell blasts. 1998 5320 5330 - 17.
Hatalski, C.G., W.F. Hickey, and W.I. Lipkin, Evolution of the immune response in the central nervous system following infection with Borna disease virus. J Neuroimmunol,1998 137 142 - 18.
Hatalski, C.G., W.F. Hickey, and W.I. Lipkin, Humoral immunity in the central nervous system of Lewis rats infected with Borna disease virus. J Neuroimmunol,1998 128 136 - 19.
Science,Hickey W. F. Kimura H. Perivascular microglial. cells of. the C. N. S. are bone. marrow-derived present antigen. in vivo. 1988 290 292 - 20.
Yamanaka, R., Dendritic-cell- and peptide-based vaccination strategies for glioma. Neurosurg Rev,2009 265 273 discussion 273. - 21.
J Natl Cancer Inst,Naumov G. N. et al. model A. of human. tumor dormancy. an angiogenic. switch from. the nonangiogenic. phenotype 2006 316 325 - 22.
[Schneider, T., et al., Increased concentrations of transforming growth factor beta1 and beta2 in the plasma of patients with glioblastoma. J Neurooncol,2006 61 65 - 23.
Nat Immunol,Sakaguchi S. Naturally arising. Foxp3-expressing C. D2 C. D. regulatory T. cells in. immunological tolerance. to self. non-self 2005 345 352 - 24.
Int J Cancer,Grauer O. M. et al. C. D. Fox P. regulatory T. cells gradually. accumulate in. gliomas during. tumor growth. efficiently suppress. antiglioma immune. responses in. vivo 2007 95 105 - 25.
Fecci, P.E., et al., Increased regulatory T-cell fraction amidst a diminished CD4 compartment explains cellular immune defects in patients with malignant glioma. Cancer Res,2006 3294 3302 - 26.
[Yamanaka, R., 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 1172 1179 - 27.
Kikuchi, T., et al., Vaccination of glioma patients with fusions of dendritic and glioma cells and recombinant human interleukin 12. J Immunother,2004 452 459 - 28.
Cancer Immunol Immunother,Kikuchi T. et al. Results of. a. phase I. clinical trial. of vaccination. of glioma. patients with. fusions of. dendritic glioma cells. 2001 337 344 - 29.
Rutkowski, S., et al., Surgery and adjuvant dendritic cell-based tumour vaccination for patients with relapsed malignant glioma, a feasibility study. Br J Cancer,2004 1656 1662 - 30.
Yajima, N., et al., Immunologic evaluation of personalized peptide vaccination for patients with advanced malignant glioma. Clin Cancer Res,2005 5900 5911 - 31.
Science,van der Bruggen P. et al. gene A. encoding an. antigen recognized. by cytolytic. T. lymphocytes on. a. human melanoma. 1991 1643 1647 - 32.
Proc Natl Acad Sci U S A,Chen Y. T. et al. testicular A. antigen aberrantly. expressed in. human cancers. detected by. autologous antibody. screening 1997 1914 1918 - 33.
Debinski, W., et al., Receptor for interleukin 13 is abundantly and specifically over-expressed in patients with glioblastoma multiforme. Int J Oncol,1999 481 486 - 34.
Fichtner-Feigl, S., et al., IL-13 signaling through the IL-13alpha2 receptor is involved in induction of TGF-beta1 production and fibrosis. Nat Med,2006 99 106 - 35.
Clin Cancer Res,Wykosky J. et al. Interleukin- receptor alpha. . Eph A. Fos-related antigen. . as molecular denominators. of high-grade. astrocytomas specific targets. for combinatorial. therapy 2008 199 208 - 36.
Das, A., et al., Expression of survivin in primary glioblastomas. J Cancer Res Clin Oncol,2002 302 306 - 37.
Chakravarti, A., et al., Quantitatively determined survivin expression levels are of prognostic value in human gliomas. J Clin Oncol,2002 1063 1068 - 38.
Shankar, S.L., et al., Survivin inhibition induces human neural tumor cell death through caspase-independent and-dependent pathways. J Neurochem,2001 426 436 - 39.
Jeon, B.G., et al., Characterization and comparison of telomere length, telomerase and reverse transcriptase activity and gene expression in human mesenchymal stem cells and cancer cells of various origins. Cell Tissue Res,2011 - 40.
Sallinen, P., et al., Increased expression of telomerase RNA component is associated with increased cell proliferation in human astrocytomas. Am J Pathol,1997 1159 1164 - 41.
DeMasters, B.K., et al., Differential telomerase expression in human primary intracranial tumors. Am J Clin Pathol,1997 548 554 - 42.
Beck, S., et al., Telomerase activity-independent function of TERT allows glioma cells to attain cancer stem cell characteristics by inducing EGFR expression. Mol Cells,2011 9 15 - 43.
Scheurer, M.E., et al., Detection of human cytomegalovirus in different histological types of gliomas. Acta Neuropathol,2008 79 86 - 44.
Clin Cancer Res,Fecci P. E. et al. Systemic-C anti. D. monoclonal antibody. administration safely. enhances immunity. in murine. glioma without. eliminating regulatory. T. cells 2006 Pt 1):4294 4305 - 45.
J Immunol,Liyanage U. K. et al. Prevalence of. regulatory T. cells is. increased in. peripheral blood. tumor microenvironment. of patients. with pancreas. or breast. adenocarcinoma 2002 2756 2761 - 46.
Elliott, L.H., W.H. Brooks, and T.L. Roszman, Cytokinetic basis for the impaired activation of lymphocytes from patients with primary intracranial tumors. J Immunol,1984 1208 1215 - 47.
Elliott, L.H., W.H. Brooks, and T.L. Roszman, Activation of immunoregulatory lymphocytes obtained from patients with malignant gliomas. J Neurosurg,1987 231 236 - 48.
J Neurosurg,Mc Vicar D. W. Davis D. F. Merchant R. E. In vitro. analysis of. the proliferative. potential of. T. cells from. patients with. brain tumor. glioma-associated immunosuppression. unrelated to. intrinsic cellular. defect 1992 251 260 - 49.
Roszman, T.L. and W.H. Brooks, Immunobiology of primary intracranial tumours. III. Demonstration of a qualitative lymphocyte abnormality in patients with primary brain tumours. Clin Exp Immunol,1980 395 402 - 50.
J Immunol,Morford L. A. et al. cell T. receptor-mediated signaling. is defective. in T. cells obtained. from patients. with primary. intracranial tumors. 1997 4415 4425 - 51.
EMBO J,de Martin R. et al. Complementary D. N. A. for human. glioblastoma-derived T. cell suppressor. factor a. novel member. of the. transforming growth. factor-beta gene. family E. M. B. 1987 3673 3677 - 52.
J.H., et al., J Exp Med,Kehrl J. H. et al. Production of. transforming growth. factor beta. by human. T. lymphocytes its potential. role in. the regulation. of T. cell growth. 1986 1037 1050 - 53.
Eur J Immunol,Zuber P. Kuppner M. C. De Tribolet N. Transforming growth. factor-beta . down-regulates-D H. L. A. antigen R. expression on. human malignant. glioma cells. 1988 1623 1626 - 54.
Proc Natl Acad Sci U S A,Torre-Amione G. et al. highly A. immunogenic tumor. transfected with. a. murine transforming. growth factor. type beta. . c. D. N. A. escapes immune. surveillance 1990 1486 1490 - 55.
Ohm, J.E. and D.P. Carbone, VEGF as a mediator of tumor-associated immunodeficiency. Immunol Res,2001 263 272 - 56.
Aoki, M., et al., Effects of vascular endothelial growth factor and E-selectin on angiogenesis in the murine metastatic RCT sarcoma. Tumour Biol,2001 239 246 - 57.
Gabrilovich, D.I., et al., Antibodies to vascular endothelial growth factor enhance the efficacy of cancer immunotherapy by improving endogenous dendritic cell function. Clin Cancer Res,1999 2963 2970 - 58.
Lien S. Lowman H. B. Therapeutic-V anti. antibodies E. G. F. Handb Exp. Pharmacol 2008 131 150 - 59.
Vredenburgh, J.J., et al., J Clin Oncol,2007 4722 4729 - 60.
Vredenburgh, J.J., et al., Phase II trial of bevacizumab and irinotecan in recurrent malignant glioma. Clin Cancer Res,2007 1253 1259 - 61.
Bao, S., et al., Stem cell-like glioma cells promote tumor angiogenesis through vascular endothelial growth factor. Cancer Res,2006 7843 7848 - 62.
J Exp Med,Seddon B. Mason D. Regulatory T. cells in. the control. of autoimmunity. the essential. role of. transforming growth. factor beta. interleukin . in the. prevention of. autoimmune thyroiditis. in rats. by peripheral. C. D4( C. D. -cells R. C. D4( C. D8( C. thymocytes 1999 279 288 - 63.
Bagavant, H., et al., Differential effect of neonatal thymectomy on systemic and organ-specific autoimmune disease. Int Immunol,2002 1397 1406 - 64.
Proc Natl Acad Sci U S A,Hori S. et al. Specificity requirements. for selection. effector functions. of C. D25+ regulatory T. cells in. anti-myelin basic. protein T. cell receptor. transgenic mice. 2002 8213 8218 - 65.
Blood,Ng W. F. et al. Human C. D4( D25( C. cells a. naturally occurring. population of. regulatory T. cells 2001 2736 2744 - 66.
J., et al., J Immunol,Ermann J. et al. D4( C. D25( C. cells T. facilitate the. induction of. T. cell anergy. 2001 4271 4275 - 67.
Cancer Res,Somasundaram R. et al. Inhibition of. cytolytic T. lymphocyte proliferation. by autologous. C. D4 D2 C. regulatory T. cells in. a. colorectal carcinoma. patient is. mediated by. transforming growth. factor-beta 2002 5267 5272 - 68.
Nat Med,Curiel T. J. et al. Specific recruitment. of regulatory. T. cells in. ovarian carcinoma. fosters immune. privilege predicts reduced. survival 2004 942 949 - 69.
J Natl Compr Canc Netw,Reardon D. A. et al. Review A. of-Targeted V. E. G. F. V. E. G. F. R. Therapeutics for. Recurrent Glioblastoma. 2011 414 427 - 70.
Clin Cancer Res,Lucio-Eterovic A. K. Piao Y. de Groot J. F. Mediators of. glioblastoma resistance. invasion during. antivascular endothelial. growth factor. therapy 2009 4589 4599 - 71.
Libermann, T.A., et al., Amplification, enhanced expression and possible rearrangement of EGF receptor gene in primary human brain tumours of glial origin. Nature,1985 144 147 - 72.
Bigner, S.H., et al., Characterization of the epidermal growth factor receptor in human glioma cell lines and xenografts. Cancer Res,1990 8017 8022 - 73.
Batra, S.K., et al., Epidermal growth factor ligand-independent, unregulated, cell-transforming potential of a naturally occurring human mutant EGFRvIII gene. Cell Growth Differ,1995 1251 1259 - 74.
Proc Natl Acad Sci U S A,Nishikawa R. et al. mutant A. epidermal growth. factor receptor. common in. human glioma. confers enhanced. tumorigenicity 1994 7727 7731 - 75.
Huang, H.S., et al., The enhanced tumorigenic activity of a mutant epidermal growth factor receptor common in human cancers is mediated by threshold levels of constitutive tyrosine phosphorylation and unattenuated signaling. J Biol Chem,1997 2927 2935 - 76.
Lammering, G., et al., Inhibition of the type III epidermal growth factor receptor variant mutant receptor by dominant-negative EGFR-CD533 enhances malignant glioma cell radiosensitivity. Clin Cancer Res,2004 6732 6743 - 77.
Cancer Res,Nagane M. et al. common A. mutant epidermal. growth factor. receptor confers. enhanced tumorigenicity. on human. glioblastoma cells. by increasing. proliferation reducing apoptosis. 1996 5079 5086 - 78.
[Montgomery, R.B., et al., Expression of oncogenic epidermal growth factor receptor family kinases induces paclitaxel resistance and alters beta-tubulin isotype expression. J Biol Chem,2000 17358 17363 - 79.
Wikstrand, C.J., et al., Monoclonal antibodies against EGFRvIII are tumor specific and react with breast and lung carcinomas and malignant gliomas. Cancer Res,1995 3140 3148 - 80.
Frederick, L., et al., Diversity and frequency of epidermal growth factor receptor mutations in human glioblastomas. Cancer Res,2000 1383 1387 - 81.
Schlegel, J., et al., Amplification of the epidermal-growth-factor-receptor gene correlates with different growth behaviour in human glioblastoma. Int J Cancer,1994 72 77 - 82.
Sugawa, N., et al., Identical splicing of aberrant epidermal growth factor receptor transcripts from amplified rearranged genes in human glioblastomas. Proc Natl Acad Sci U S A,1990 8602 8606 - 83.
Wikstrand, C.J., et al., Cell surface localization and density of the tumor-associated variant of the epidermal growth factor receptor, EGFRvIII. Cancer Res,1997 4130 4140 - 84.
Humphrey, P.A., et al., Anti-synthetic peptide antibody reacting at the fusion junction of deletion-mutant epidermal growth factor receptors in human glioblastoma. Proc Natl Acad Sci U S A,1990 4207 4211 - 85.
Sampson, J.H., et al., Unarmed, tumor-specific monoclonal antibody effectively treats brain tumors. Proc Natl Acad Sci U S A,2000 7503 7508 - 86.
Reist, C.J., et al., Tumor-specific anti-epidermal growth factor receptor variant III monoclonal antibodies: use of the tyramine-cellobiose radioiodination method enhances cellular retention and uptake in tumor xenografts. Cancer Res,1995 4375 4382 - 87.
Lorimer, I.A., et al., Immunotoxins that target an oncogenic mutant epidermal growth factor receptor expressed in human tumors. Clin Cancer Res,1995 859 864 - 88.
Archer, G.E., et al., Regional treatment of epidermal growth factor receptor vIII-expressing neoplastic meningitis with a single-chain immunotoxin, MR-1. Clin Cancer Res,1999 2646 2652 - 89.
Reist, C.J., et al., In vitro and in vivo behavior of radiolabeled chimeric anti-EGFRvIII monoclonal antibody: comparison with its murine parent. Nucl Med Biol,1997 639 647 - 90.
Reist, C.J., et al., Improved targeting of an anti-epidermal growth factor receptor variant III monoclonal antibody in tumor xenografts after labeling using N-succinimidyl 5-iodo-3-pyridinecarboxylate. Cancer Res,1997 1510 1515 - 91.
Reist, C.J., et al., Astatine-211 labeling of internalizing anti-EGFRvIII monoclonal antibody using N-succinimidyl 5-[211At]astato-3-pyridinecarboxylate. Nucl Med Biol,1999 405 411 - 92.
Neurosurgery,Faillot T. et al. phase A. study I. of an. anti-epidermal growth. factor receptor. monoclonal antibody. for the. treatment of. malignant gliomas. 1996 478 483 - 93.
Stragliotto, G., et al., Multiple infusions of anti-epidermal growth factor receptor (EGFR) monoclonal antibody (EMD 55,900) in patients with recurrent malignant gliomas. Eur J Cancer,1996 A(4):636 640 - 94.
Purev, E., et al., Immune responses of breast cancer patients to mutated epidermal growth factor receptor (EGF-RvIII, Delta EGF-R, and de2-7 EGF-R). J Immunol,2004 6472 6480 - 95.
Sampson, J.H., et al., J Clin Oncol,2010 4722 4729 - 96.
Sampson, J.H., et al., Greater chemotherapy-induced lymphopenia enhances tumor-specific immune responses that eliminate EGFRvIII-expressing tumor cells in patients with glioblastoma. Neuro Oncol,2011 324 333 - 97.
Glia,Kurpad S. N. et al. Tumor antigens. in astrocytic. gliomas 1995 244 256 - 98.
J Clin Oncol,Bigner D. D. et al. Iodine-131-labeled antitenascin. monoclonal antibody. . treatment C. of patients. with recurrent. malignant gliomas. phase I. trial results. 1998 2202 2212 - 99.
Clin Cancer Res,Brown M. T. et al. Intrathecal 1. I-labeled antitenascin. monoclonal antibody. . treatment C. of patients. with leptomeningeal. neoplasms or. primary brain. tumor resection. cavities with. subarachnoid communication. phase I. trial results. 1996 963 972 - 100.
J Neurooncol,Bigner D. D. et al. Phase I. studies of. treatment of. malignant gliomas. neoplastic meningitis. with 1. I-radiolabeled monoclonal. antibodies anti-tenascin. . C. anti-chondroitin proteoglycan. sulfate Me1-. (ab’)2--a F. preliminary report. 1995 109 122 - 101.
Reardon, D.A., et al., Salvage radioimmunotherapy with murine iodine-131-labeled antitenascin monoclonal antibody 81C6 for patients with recurrent primary and metastatic malignant brain tumors: phase II study results. J Clin Oncol,2006 115 122 - 102.
Reardon, D.A., et al., Phase II trial of murine (131)I-labeled antitenascin monoclonal antibody 81C6 administered into surgically created resection cavities of patients with newly diagnosed malignant gliomas. J Clin Oncol,2002 1389 1397 - 103.
Neuro Oncol,Reardon D. A. et al. pilot A. study 1. I-antitenascin monoclonal. antibody 81c. to deliver. a. 4. Gy resection. cavity boost. 2008 182 189 - 104.
Turkington, T.G., et al., Measuring astatine-211 distributions with SPECT. Phys Med Biol, 1993 1121 1130 - 105.
Zalutsky, M.R., et al., Clinical experience with alpha-particle emitting 211At: treatment of recurrent brain tumor patients with 211At-labeled chimeric antitenascin monoclonal antibody 81C6. J Nucl Med,2008 30 38 - 106.
Nature,Steinman R. M. Banchereau J. Taking dendritic. cells into. medicine 2007 419 426 - 107.
Melief, C.J., Cancer immunotherapy by dendritic cells. Immunity,2008 372 383 - 108.
Steinman R. M. Hawiger D. Nussenzweig M. C. Tolerogenic dendritic. cells Annu. Rev Immunol. 2003 685 711 - 109.
Blood,Dhodapkar M. V. Steinman R. M. Antigen-bearing immature. dendritic cells. induce peptide-specific. C. D8( regulatory T. cells in. vivo in. humans 2002 174 177 - 110.
Immunity,Wakkach A. et al. Characterization of. dendritic cells. that induce. tolerance regulatory T. cell . differentiation in. vivo 2003 605 617 - 111.
J. Banchereau, J Immunol,Palucka K. Ueno H. Banchereau J. Recent developments. in cancer. vaccines 2011 1325 1331 - 112.
Sampson, J.H., et al., Intracerebral infusion of an EGFR-targeted toxin in recurrent malignant brain tumors. Neuro Oncol,2008 320 329 - 113.
Zaba, L.C., et al., Normal human dermis contains distinct populations of CD11c+BDCA-1+ dendritic cells and CD163+FXIIIA+ macrophages. J Clin Invest,2007 2517 2525 - 114.
Klechevsky, E., et al., Functional specializations of human epidermal Langerhans cells and CD14+ dermal dendritic cells. Immunity,2008 497 510 - 115.
Celluzzi, C.M. and L.D. Falo, Jr., J Invest Dermatol,1997 716 720 - 116.
Palucka, K., et al., Dendritic cells and immunity against cancer. J Intern Med,2011 64 73 - 117.
Lapenta, C., et al., Potent immune response against HIV-1 and protection from virus challenge in hu-PBL-SCID mice immunized with inactivated virus-pulsed dendritic cells generated in the presence of IFN-alpha. J Exp Med,2003 361 367 - 118.
Eur J Immunol,Dubsky P. et al. L-15-induced I. human D. C. efficiently prime. melanoma-specific naive. C. D. cells T. to differentiate. into C. T. L. 2007 1678 1690 - 119.
Liau, L.M., 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 5515 5525 - 120.
Yamanaka, R., et al., Clin Cancer Res,2005 4160 4167 - 121.
Yu, J.S., et al., Vaccination with tumor lysate-pulsed dendritic cells elicits antigen-specific, cytotoxic T-cells in patients with malignant glioma. Cancer Res,2004 4973 4979 - 122.
Yu, J.S., et al., Vaccination of malignant glioma patients with peptide-pulsed dendritic cells elicits systemic cytotoxicity and intracranial T-cell infiltration. Cancer Res,2001 842 847 - 123.
Morse, M.A., et al., Migration of human dendritic cells after injection in patients with metastatic malignancies. Cancer Res,1999 56 58 - 124.
Barratt-Boyes, S.M., et al., Maturation and trafficking of monocyte-derived dendritic cells in monkeys: implications for dendritic cell-based vaccines. J Immunol,2000 2487 2495 - 125.
Immunol Rev,Mitchell D. A. Fecci P. E. Sampson J. H. Immunotherapy of. malignant brain. tumors 2008 70 100 - 126.
De Vleeschouwer, S., et al., Postoperative adjuvant dendritic cell-based immunotherapy in patients with relapsed glioblastoma multiforme. Clin Cancer Res,2008 3098 3104 - 127.
Wheeler, C.J., et al., Vaccination elicits correlated immune and clinical responses in glioblastoma multiforme patients. Cancer Res,2008 5955 5964 - 128.
Prins, R.M., et al., Gene expression profile correlates with T-cell infiltration and relative survival in glioblastoma patients vaccinated with dendritic cell immunotherapy. Clin Cancer Res,2011 1603 1615 - 129.
J Immunol,Prins R. M. et al. The T. L. R. agonist imiquimod. enhances dendritic. cell survival. promotes tumor. antigen-specific T. cell priming. relation to. central nervous. system antitumor. immunity 2006 157 164 - 130.
Kundig, T.M., et al., On the role of antigen in maintaining cytotoxic T-cell memory. Proc Natl Acad Sci U S A,1996 9716 9723 - 131.
Matzinger, P., An innate sense of danger. Semin Immunol,1998 399 415 - 132.
Cancer Res,Boczkowski D. et al. Induction of. tumor immunity. cytotoxic T. lymphocyte responses. using dendritic. cells transfected. with messenger. R. N. A. amplified from. tumor cells. 2000 1028 1034 - 133.
Heiser, A., et al., Human dendritic cells transfected with RNA encoding prostate-specific antigen stimulate prostate-specific CTL responses in vitro. J Immunol,2000 5508 5514 - 134.
Wintterle, S., et al., Expression of the B7-related molecule B7-H1 by glioma cells: a potential mechanism of immune paralysis. Cancer Res,2003 7462 7467 - 135.
Dong, H., et al., Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med,2002 793 800 - 136.
Parsa, A.T., et al., Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma. Nat Med,2007 84 88 - 137.
Kugler, A., et al., Regression of human metastatic renal cell carcinoma after vaccination with tumor cell-dendritic cell hybrids. Nat Med,2000 332 336 - 138.
Cancer Immunol Immunother,Blank C. Mackensen A. Contribution of. the-L P. D. P. D. pathway to. T-cell exhaustion. an update. on implications. for chronic. infections tumor evasion. 2007 739 745 - 139.
Trends Mol Med,Yao S. Chen L. Reviving exhausted. T. lymphocytes during. chronic virus. infection by. B. blockade H. 2006 244 246 - 140.
Caruso, D.A., et al., Results of a phase 1 study utilizing monocyte-derived dendritic cells pulsed with tumor RNA in children and young adults with brain cancer. Neuro Oncol,2004 236 246 - 141.
Thompson, R.H., et al., B7-H1 glycoprotein blockade: a novel strategy to enhance immunotherapy in patients with renal cell carcinoma. Urology,2005 Suppl):10 14 - 142.
J Neurooncol,Kirsch M. Fischer H. Schackert G. Activated monocytes. kill malignant. brain tumor. cells in. vitro 1994 35 45 - 143.
Wahl, S.M., et al., Transforming growth factor-beta is a potent immunosuppressive agent that inhibits IL-1-dependent lymphocyte proliferation. J Immunol,1988 3026 3032 - 144.
Rook, A.H., et al., Effects of transforming growth factor beta on the functions of natural killer cells: depressed cytolytic activity and blunting of interferon responsiveness. J Immunol,1986 3916 3920 - 145.
Strome, S.E., et al., B7-H1 blockade augments adoptive T-cell immunotherapy for squamous cell carcinoma. Cancer Res,2003 6501 6505 - 146.
Curiel, T.J., et al., Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat Med,2003 562 567 - 147.
Suda, T., et al., Molecular cloning and expression of the Fas ligand, a novel member of the tumor necrosis factor family. Cell,1993 1169 1178 - 148.
Steinbok, P., et al., Intratumoral autologous mononuclear cells in the treatment of recurrent glioblastoma multiforme. A phase 1 (toxicity) study. J Neurooncol,1984 147 151 - 149.
Cancer,Young H. Kaplan A. Regelson W. Immunotherapy with. autologous white. cell infusions. ("lymphocytes in the. treatment of. recurrrent glioblastoma. multiforme a. preliminary report. 1977 1037 1044 - 150.
Hayes, R.L., et al., Improved long term survival after intracavitary interleukin-2 and lymphokine-activated killer cells for adults with recurrent malignant glioma. Cancer,1995 840 852 - 151.
Blancher, A., et al., Local immunotherapy of recurrent glioblastoma multiforme by intracerebral perfusion of interleukin-2 and LAK cells. Eur Cytokine Netw,1993 331 341 - 152.
Cancer Res,Jacobs S. K. et al. Interleukin or autologous. lymphokine-activated killer. cell treatment. of malignant. glioma phase. I. trial 1986 Pt 2):2101 2104 - 153.
Enhancement of MAK lytic activity and cytokine production by PHA and clinical use of PHA. J Neurooncol,Jeffes E. W. et al. Therapy of. recurrent high. grade gliomas. with surgery. autologous mitogen. activated I. L. stimulated killer. . M. A. K. lymphocytes I. 1993 141 155 - 154.
Ingram, M., et al., Immunotherapy for recurrent malignant glioma: an interim report on survival. Neurol Res,1990 265 273 - 155.
Quattrocchi, K.B., et al., Pilot study of local autologous tumor infiltrating lymphocytes for the treatment of recurrent malignant gliomas. J Neurooncol,1999 141 157 - 156.
Jpn J Cancer Res,Tsurushima H. et al. Reduction of. end-stage malignant. glioma by. injection with. autologous cytotoxic. T. lymphocytes 1999 536 545 - 157.
J Neurooncol,Kitahara T. et al. Establishment of. interleukin . dependent cytotoxic. T. lymphocyte cell. line specific. for autologous. brain tumor. its intracranial. administration for. therapy of. the tumor. 1987 329 336 - 158.
The Fred Hutchinson Cancer Research Center and the University of Washington School of Medicine, Department of Medicine, Division of Oncology. Hum Gene Ther,Riddell S. R. et al. Phase I. study of. cellular adoptive. immunotherapy using. genetically modified. C. D. V-specific H. I. cells T. for H. I. V. seropositive patients. undergoing allogeneic. bone marrow. transplant 1992 319 338 - 159.
Papadopoulos, E.B., et al., Infusions of donor leukocytes to treat Epstein-Barr virus-associated lymphoproliferative disorders after allogeneic bone marrow transplantation. N Engl J Med,1994 1185 1191 - 160.
Kolb, H.J., et al., Donor leukocyte transfusions for treatment of recurrent chronic myelogenous leukemia in marrow transplant patients. Blood,1990 2462 2465 - 161.
Brooks, W.H., et al., Relationship of lymphocyte invasion and survival of brain tumor patients. Ann Neurol,1978 219 224 - 162.
Incidence, fate, and relevance to prognosis in 228 operated cases. J Neurosurg,Palma L. Di Lorenzo N. Guidetti B. Lymphocytic infiltrates. in primary. glioblastomas recidivous gliomas. 1978 854 861 - 163.
Rosenberg, S. A., Ann Surg,1988 121 135 - 164.
Jacobs, S.K., et al., Interleukin-2 and autologous lymphokine-activated killer cells in the treatment of malignant glioma. Preliminary report. J Neurosurg,1986 743 749 - 165.
Merchant, R.E., et al., Intralesional infusion of lymphokine-activated killer (LAK) cells and recombinant interleukin-2 (rIL-2) for the treatment of patients with malignant brain tumor. Neurosurgery,1988 725 732 - 166.
Blacklock, J.B. and E.A. Grimm, Lymphokine-activated killer lymphocytes: LAK and interleukin-2 in the treatment of malignancies of the central nervous system. Immunol Ser,1989 93 99 - 167.
Lillehei, K.O., et al., Long-term follow-up of patients with recurrent malignant gliomas treated with adjuvant adoptive immunotherapy. Neurosurgery,1991 16 23 - 168.
Dillman, R.O., et al., Intracavitary placement of autologous lymphokine-activated killer (LAK) cells after resection of recurrent glioblastoma. J Immunother,2004 398 404 - 169.
Dudley, M.E., et al., Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science,2002 850 854 - 170.
Cancer Immunol Immunother,Kruse C. A. et al. Treatment of. recurrent glioma. with intracavitary. alloreactive cytotoxic. T. lymphocytes interleukin- 1997 77 87 - 171.
Kawada, M., et al., Vaccination of fusion cells of rat dendritic and carcinoma cells prevents tumor growth in vivo. Int J Cancer,2003 520 526 - 172.
Komata, T., et al., Telomerase as a therapeutic target for malignant gliomas. Oncogene,2002 656 663 - 173.
Sampson, J.H. and D.A. Mitchell, Is Cytomegalovirus a Therapeutic Target in Glioblastoma? Clin Cancer Res,2011 . - 174.
J Immunol,Itoh M. et al. Thymus autoimmunity production. of C. D2 C. D. naturally anergic. suppressive T. as cells a. key function. of the. thymus in. maintaining immunologic. self-tolerance 1999 5317 5326 - 175.
Bruder, C.E., et al., High resolution deletion analysis of constitutional DNA from neurofibromatosis type 2 (NF2) patients using microarray-CGH. Hum Mol Genet,2001 271 282 - 176.
J Immunol,Pacholczyk R. Kraj P. Ignatowicz L. Peptide specificity. of thymic. selection of. C. D. D2 C. cells T. 2002 613 620 - 177.
Clin Cancer Res,Wolf A. M. et al. Increase of. regulatory T. cells in. the peripheral. blood of. cancer patients. 2003 606 612 - 178.
Clin Cancer Res,Ichihara F. et al. Increased populations. of regulatory. T. cells in. peripheral blood. tumor-infiltrating lymphocytes. in patients. with gastric. esophageal cancers. 2003 4404 4408 - 179.
Cancer,Sasada T. et al. C. D. D2 C. regulatory T. cells in. patients with. gastrointestinal malignancies. possible involvement. of regulatory. T. cells in. disease progression. 2003 1089 1099 - 180.
Cancer Res,Woo E. Y. et al. Regulatory C. D4( D25( C. cells T. in tumors. from patients. with early-stage. non-small cell. lung cancer. late-stage ovarian. cancer 2001 4766 4772 - 181.
Cell,Kappler J. W. Roehm N. Marrack P. cell T. tolerance by. clonal elimination. in the. thymus 1987 273 280 - 182.
Kisielow, P., et al., Tolerance in T-cell-receptor transgenic mice involves deletion of nonmature CD4+8+ thymocytes. Nature,1988 742 746 - 183.
Cancer Lett,Schmitz M. et al. Identification of. a. naturally processed. T. cell epitope. derived from. the glioma-associated. protein S. O. X1 2007 331 336 - 184.
Immunol Rev,Wekerle H. et al. The shaping. of the. brain-specific T. lymphocyte repertoire. in the. thymus 1996 231 243 - 185.
J Exp Med,Asano M. et al. Autoimmune disease. as consequence a. of developmental. abnormality of. a. T. cell subpopulation. 1996 387 396 - 186.
Rosenberg, S.A., et al., Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Nat Rev Cancer,2008 299 308 - 187.
J Clin Invest,Wrzesinski C. et al. Hematopoietic stem. cells promote. the expansion. function of. adoptively transferred. antitumor C. D. T. cells 2007 492 501 - 188.
Morgan, R.A., M.E. Dudley, and S.A. Rosenberg, Adoptive cell therapy: genetic modification to redirect effector cell specificity. Cancer J,2010 336 341