Abstract
With the recent approval by the FDA of an immunotherapy for prostate cancer and another positive immunotherapy trial in melanoma, immunotherapy may finally be coming of age. So what will it take for it to become part of the standard treatment for glioblastoma? To put this question into perspective, we summarize critical background information in neuro-immunology, address immunotherapy clinical trial design, and discuss a number of extrinsic factors that will impact the development of immunotherapy in neuro-oncology.
Keywords: clinical trial, glioblastoma multiforme, immunotherapy, malignant gliomas
Immune Privilege
The failure of immunotherapeutics to exert an effect against tumors within the brain has been attributed to “immunological privilege”1 secondary to the absence of a lymphatic drainage system within the brain, the presence of the blood–brain barrier (BBB), and a paucity of resident specialized antigen-presenting cells (APCs) within the CNS.2,3 However, all these premises have now been substantially discounted. For example, cerebrospinal fluid (CSF) has been shown to drain via the Virchow–Robbin spaces to the deep cervical lymphatics4 via perivascular sheaths and through the nasal submucosa.5–7 Antigens within the CNS enter the cervical lymph nodes by these routes8 and result in immune activation with a distinct hierarchy.9 This hierarchy is characterized by strong antibody responses and priming of cytotoxic T cell responses but an absence of delayed-type hypersensitivity (DTH) responses, with a skewing toward a nontumor Th2 phenotype.6,8–10 Although naïve T cells are not found within the CNS, T cells and antibodies have access to antigens within the CNS,11–13 indicating that the BBB does not form an absolute barrier to immune responses. Activated T cells are permitted to patrol the CNS in an antigen-independent and apparently unrestricted manner14 and return to the systemic circulation. These cells exit through the cribriform plate and reach the nasal mucosa and, eventually, cervical lymph nodes.15 Evidence suggests that T cells that encounter their cognate antigen are retained within the CNS,16 but some studies suggest that they do not proliferate and instead undergo apoptosis.17,18 Other studies have shown the proliferation of antigen-specific T cells, specifically within tumors, and differentiation into cells with enhanced effector function.16 Microglia and macrophages have been shown to act as resident APCs within the CNS.19 Dendritic cells (DCs) are present in both the choroid plexus20 and meninges.21 CNS microglia,22 with the phenotypic and functional characteristics of both macrophages and DCs,23,24 express class II antigens and T cell costimulatory molecules24–26 that are capable of antigen presentation when not associated with tumors. Astrocytes, though capable of antigen presentation, are poor APCs and probably do not play a primary role in immune activation.17
Based largely on the low globulin protein concentration within the CSF, it was generally believed that antibodies do not penetrate the BBB. However, antibodies do rapidly accumulate within the CSF and brain parenchyma after passive or active peripheral immunization in experimental animals18 and are distributed throughout the CNS according to kinetics similar to those in other peripheral organs, albeit at a ratio of ∼0.1%–5% of the titer of antibodies found in the serum.27–30 Despite this, it remains unclear as to what levels of antibody are sufficient to mediate effector functions in the brain, including antitumor effects, and how binding kinetics and antigen distribution affect these parameters. Soon after the approval of trastuzumab for HER2-positive breast cancer, concern arose over an apparent increase in CNS metastases,31,32 particularly in the context of excellent systemic control. This was taken as evidence that antibodies like trastuzumab cannot cross the BBB at levels sufficient to have a therapeutic effect, even in the context of metastatic intraparenchymal tumor or leptomeningeal disease. However, the cause of this apparent increase in brain metastases in patients with HER2-positive breast cancer is probably multifactorial. Subsequent studies have shown that trastuzumab does enter the CSF, but at significantly lower concentrations (∼420:1), even in the context of whole-brain radiation therapy, which is thought to disrupt the BBB (76:1), or leptomeningeal disease (<49:1).29,30 Furthermore, PET imaging has recently shown directly that trastuzumab can penetrate brain metastases.33 However, what has also been shown is that CNS metastases from breast cancer remain HER2 positive; therefore, it is not clear what amount of antibody that penetrates the CNS is sufficient to manifest an antitumor effect. Cumulatively, these data indicate that tumors in the CNS should not be considered “off-limits” to immunotherapy and that the therapeutic nihilism surrounding the application of immunotherapy to primary and metastatic tumors of the brain needs to be eliminated.
Immunosuppression
Patients with cancer, and especially those with malignant gliomas, have a variety of heterogeneous, redundant mechanisms that contribute to their overall state of immune suppression, and these mechanisms serve as a barrier to effective immunotherapy. Generalized manifestations of immune impairment in these patients include low peripheral lymphocyte counts, reduced DTH reactions to recall antigens, impaired mitogen-induced blastogenic responses by peripheral blood mononuclear cells (PBMCs), and increased numbers of regulatory T cells (Tregs; reviewed in Dey et al.34). Primed CD8+ cytotoxic T cells gain CNS access14,35; however, the lack of tumor eradication indicates that the T cells are functionally impaired. This has been confirmed with ex vivo studies demonstrating a lack of effector/activated T cells in the glioma microevironment.36 More specifically, adaptive immune responses are noticeably deficient, with diminished responsiveness of peripheral T cells associated with impaired early transmembrane signaling through the T cell receptor/CD3 complex. In addition, reduced immunoglobulin synthesis by B cells in vitro from the peripheral blood of patients with intracranial tumors appears to be related to diminished T-helper cell activity. Many cancers, including gliomas, secrete factors such as prostaglandin E2, interleukin (IL)-10, vascular endothelial growth factor, and transforming growth factor (TGF)–β that are capable of suppressing cytotoxic responses of T cells against tumor targets, downregulating major histocompatibility complex (MHC) expression, suppressing T cell proliferation,37–39 and inhibiting the maturation of DCs.40 The absence or low expression of costimulatory molecules within the CNS36 also gives an immune escape advantage to cancer cells because costimulatory signals are essential for differentiation of functional tumor-specific CD8+ T-effector cells.41–44 Furthermore, the expression of costimulatory inhibitory molecules like B7-H1 that are expressed in malignant gliomas (especially with PTEN gene loss) can further inhibit immune responses.45
Many studies have demonstrated that Tregs are responsible for inhibition of tumor reactive effector T cells, and the elimination of Tregs by any of several different strategies successfully enhances antitumor immunity mostly in murine models.46–50 Similarly, CD4+CD25+FoxP3+ Treg-mediated suppression has also been demonstrated in several human cancers with increased numbers of these cells.51–54 Collectively, these data indicate that Tregs cannot only inhibit the initial systemic immune activation but also prevent the effector responses in the tumor microenvironment. Increasingly, other immune populations such as immunosuppressive myeloid cells55 and M2 macrophages56,57 within the tumor microenvironment are being shown to participate and mediate tumor immunosuppression.
A variety of investigators have also shown that cancer stem cells suppress immune response,58–61 indicating that this is probably a generalized property of pluripotent stem cells. Distinct pathways and mechanisms that are used by the cancer stem cells, if they exist, will need to be clarified to specifically target cancer stem cell–mediated immunosuppression. It may be possible to vaccinate against cancer stem cells through the use of immunologic manipulations that enable the generated effector responses to overwhelm the immunosuppressive capacity of the tumor.62–64 Nevertheless, the grim reality is that recurrence and persistence are hallmark features of gliomas, and in most circumstances, the intrinsic immune system of the patient, unaided, is unable to eradicate the cancer stem cells that are the progenitors that give rise to recurrence and progression. Thus, immunosuppression predominates in patients with high-grade gliomas and poses barriers for successful antitumor immunotherapy.
A rudimentary way to overcome this tumor-mediated immunosuppression is by attempting to resect as much of the tumor as is feasibly possible—an approach that has been used in several recent clinical trials (Table 1). This also provides the opportunity to minimize the patient's dependence on immunosuppressive steroids, a possible confounding factor during active immunotherapeutic approaches. However, not all patients have disease that is amenable to surgery, and thus alternative approaches that can target key molecular hubs that mediate multiple mechanisms of immunosuppression need to be identified and targeted.
Table 1.
Agent delivered/site | Phase | Sponsor or centers involved | Results |
---|---|---|---|
PEP-3-KLH + GM-CSF (ACTIVATE)/systemic120 | II | Duke University Medical Center/The University of Texas MD Anderson Cancer Center/Celldex | Median survival time = 2.4 years; newly diagnosed; n = 23 |
PEP-3-KLH + GM-CSF with TMZ (ACT II)/systemic121 | II | Duke University Medical Center/The University of Texas MD Anderson Cancer Center/Celldex/Pfizer | Median survival time = 1.9 years; newly diagnosed; n = 21 |
DCs + PEP-3-KLH/systemic109 | I/II | Duke University Medical Center | Median survival time = 1.8 years; newly diagnosed; n = 14 |
Personalized peptide vaccines (4)/systemic122 | I | Nigata University | Median survival time = 1.7 years; recurrent GBM; n = 17 |
DCs + CMV systemic123 | II | Duke University Medical Center | Median survival time not reached yet but will exceed 1.6 years; newly diagnosed GBM; n = 13 |
DCs + autologous tumor lysates124 | II | University of Leuven and Wurzburg | Median survival time from relapse = 0.8 years; recurrent GBM; n = 56 |
DCs + acid-eluted tumor peptides/systemic125 | I/II | Cedars Sinai Medical Center | Median survival time = 1.3 years; newly diagnosed GBM; n = 7 |
DCs + tumor homogenate/systemic108 | II | Cedars Sinai Medical Center | Median survival time = 1.8 years for immune responders vs 1.2 for nonimmune responders; newly diagnosed GBM; n = 11; median survival time = 1.6 years for immune responders vs 1.1 for nonimmune responders; recurrent GBM; n = 21 |
DCs + acid-eluted tumor peptides/systemic126 | I | UCLA | Median survival time = 2.0 years; newly diagnosed and recurrent GBM patients; n = 12 |
Autologous tumor cells with TGF-β2 antisense/systemic127 | I | Advanced Biotherapies/NovaRx | Median survival time = 1.4 years; progressing GBM patients at enrollment; n = 6 |
Poly-ICLC/systemic128 | II | North American Brain Tumor Coalition | Median survival time = 1.4 years; newly diagnosed GBM patients; n = 30 |
Autologous whole tumor with GM-CSF and adoptive transfer of CD3-activated lymphocytes/systemic129 | II | Tvax Biomedical | Median survival time = 1.0 years; recurrent malignant glioma; n = 19 |
KLH, keyhole limpet hemocyanin; poly-ICLC, polyinosinic-polycytidylic acid stabilized with polylysine and carboxymethylcellulose; GM-CSF, granulocyte-macrophage colony–stimulating factor.
The signal transducer and activator of transcription 3 (STAT3) has been shown to be a potent regulator of anti-inflammatory responses by suppressing innate65–68 and adaptive immunity.69 The STAT3 pathway becomes constitutively active in diverse tumor-infiltrating immune cells,69 markedly impairing their effector responses.69,70 STAT3 also increases the functional activity of the immunosuppressive Tregs.71–73 Furthermore, we have recently demonstrated that the cancer stem cells are dependent on the STAT3 pathway in mediating immunosuppression that can be reversed with p-STAT3 inhibitors,60 indicating that this pathway is a molecular hub of tumor-mediated immunosuppression.
Targeting these molecular hubs is a paradigm shift from previous approaches that attempted to overwhelm the tumor with effector responses by now focusing therapeutic attention on controlling tumor-mediated immune suppression in a comprehensive, global fashion and perhaps allowing intrinsic recognition of tumor-associated antigen (TAA) and tumor-specific antigen (TSA). Interestingly, adult malignant gliomas appear to express more TAA and TSA than gliomas within pediatric patients,74 likely related to better immunologic reactivity (ie, less immune suppression) in the pediatric patient relative to the adult75 and makes the use of agents that can profoundly control tumor-mediated immune suppression therapeutically compelling. Thus, if we are going to be successful in treating patients with glioblastoma multiforme (GBM) with immunotherapy, we will also have to carefully consider and combat the matrix of immunosuppressive mechanisms operating in these patients.
Clinical Trial Design
With immunologically based therapeutics, a maximum tolerated dose cannot be identified in most cases. This is especially true with biologic agents in which there is a limitation to the amount of “drug” that can be feasibly generated. Thus, in many cases, what can be obtained during a phase I clinical trial of an immunotherapeutic is a maximum feasible dose. Furthermore, for noncytotoxic agents and most immunotherapeutics, efficacy76 and toxicity77 are often not clearly doserelated. Added to this is the practical matter that the chances of dose escalation are very high with standard 3 + 3 designs, even when the true rate of toxicity is quite high. Shown below is the probability of escalating from one dose to another as a function of the true unknown toxicity rate in a standard 3 + 3 trial:
True toxicity rate (%) | 5 | 10 | 20 | 30 | 40 | 50 |
Probability of dose escalation | 0.97 | 0.91 | 0.71 | 0.49 | 0.31 | 0.17 |
As can be seen from this chart, there is a >90% probability of further dose escalation even when the dose-limiting toxicity rate is as high as 10% in a 3 + 3 trial design. These toxicity rates have seldom been seen within the context of cancer immunotherapeutics. Thus, in the context of clinical trials of immunotherapeutics, there are little data to support a traditional phase I dose-escalation approach based on toxicity assessment alone.77
An alternative goal of “dose-escalation” immunotherapeutic trials has been to determine the most effective dose. These studies usually list immune response as a primary endpoint because most investigators realize that the variability inherent in survival endpoints prohibits obtaining the answer to this question with a reasonable number of patients. Although a laudable goal, this is also often ill-conceived because of the variability in immune responses seen among patients as well as the small magnitude of the responses, which makes these assessments prohibitive in early-phase trials as well. For example, consider a clinical trial in which even a fairly dramatic doubling of immune response is sought between groups given different doses of a vaccine. Suppose the expected mean immune response in Group 1 is 1.0 U and that in Group 2 is 2.0 U. What sample size is required to demonstrate this difference? Consider the 4 following scenarios that assume a 2-tailed test conducted at the 0.05 level of significance with 80% power:
Group | Mean response (U) | Standard deviation |
|||
---|---|---|---|---|---|
Scenario #1 | Scenario #2 | Scenario #3 | Scenario #4 | ||
One | 1.0 | 0.1 | 0.5 | 1.0 | 2.0 |
Two | 2.0 | 0.2 | 1.0 | 2.0 | 4.0 |
Sample size | 6 | 24 | 82 | 318 |
Few (if any) immunotherapy studies demonstrate a doubling in mean immune response, and fewer still have such consistent responses that variability is reduced to the levels shown in the above table. As a result, large cohorts of patients will be required at each dose to obtain meaningful data that are sufficiently powered. As such, we do not recommend that the immunotherapeutic clinical trials be devised to detect differences in immune response with different doses of an immunotherapeutic.
Most of the phase II immunotherapeutic clinical trials conducted to date typically enroll small numbers of patients with unique eligibility criteria (Table 1) that preclude robust analysis of confounding prognostic variables because of insufficient statistical power and the lack of robust databases such as those that are maintained by the FDA for other diseases. Although comparisons with such databases using a historical cohort matched to enrollment criteria and prognostic variables may have some value, in the last 10 years, in part owing to the introduction of new agents and our more aggressive care of this patient population, the outcome for patients with GBM has rapidly shifted. For example, in the definitive clinical trial supporting the efficacy of temozolomide (TMZ) in patients newly diagnosed with GBM, the median survival time was initially reported to be 15 months.78 However, more contemporaneous clinical trials evaluating the efficacy of TMZ have demonstrated a median survival interval of 18.2 months.79 Although one could argue that these differences may be attributed to subtle differences in the timing of administration of the TMZ (concomitant with and after radiation therapy vs strictly after radiation therapy), these differences probably reflect changes in the treatment regimens of these patients, especially upon tumor recurrence. The survival benefit of more intensive medical intervention is further supported by an analysis of the median survival time of GBM patients who, based only on the criterion of being enrolled in a clinical trial regardless of the treatment agents, had a median survival time of 19.680 to 21 months.81 Thus, because the historical cohort consists of retrospective data, its use as a comparative population may not reflect current treatment responses and could convey a false sense of response unless the databases are large, homogeneous, and tightly regulated. We therefore recommend that given the recent shift in the survival time of GBM patients, randomization to a control standard-of-care arm (or an equivalent) is essential during phase II testing.
It is apparent that immunotherapy has been more successful in other tumors than GBM. For example, 2 immunotherapeutic agents, interferon (IFN)-α and IL-2, have long been approved by the FDA for stages II and III melanoma patients who had relatively low malignancy burdens based on the response rates of 10%–33% and prolongations in survival.82–85 More recently, immunotherapy has been shown to be efficacious even in advanced prostate cancer86 and unresectable stage III and IV melanoma,87 indicating that significant tumor burden can be overcome using immunotherapy approaches. What remains unclear is whether the degree or types of operational mechanisms of immune suppression are fundamentally different between malignant gliomas and other types of malignancy. As we move forward in the design of immunotherapeutic clinical trials, we recommend stratifying our patients based on the amount of residual disease and on the operational mechanisms of immunosuppression that are occurring. For example, it would seem appropriate to stratify patients according to the Treg fraction53,54,88 for clinical trials testing anti-Treg approaches.89 This would serve to identify those patients likely to benefit from those agents rather than patients who are relying on other mechanisms of immune suppression. Furthermore, Parsa et al.45 have suggested that patients who have lost tumor suppressor PTEN function and thus have upregulated B7-H1 may not be suitable candidates for active immunotherapeutic approaches at all. In the future, factors that may predict immunotherapeutic resistance—such as B7-H1 expression, Treg level, PTEN deletion, STAT3 phosphorylation, and CD133 expression—could be used as part of an immunosuppressive genetic signature90–93 to stratify patients enrolling in immunotherapeutic clinical trials. Thus, we recommend that in future immunotherapeutic clinical trials, patients be stratified based on residual disease and that the evaluation of markers that may reflect immunotherapeutic resistance (such as B7-H1, Tregs, PTEN, p-STAT3, and CD133 expression) be included in the context of a secondary endpoint or as stratification variables in the trial design.
Immune response monitoring
To date, no T cell–based immune response measure has been universally validated in cancer immunotherapy (Table 2).94–100 This is partly because of the lack of standardization or even definitive agreement on prioritization of these assays. It is also likely that evaluating a single immune cell population will be insufficient because antitumor immune responses are probably an orchestrated effort among a variety of immune cell populations that are not captured in popular rudimentary assays. Attempts to resolve this issue have included ascertaining polyvalent immune responses using multiparameter flow analysis. These assays, while accounting for more global immune responses, may also still reflect the functional status or overall immune responsive nature of a subset of patients and only in a specific compartment at a specific time.
Table 2.
Immune assay | Advantages | Disadvantages |
---|---|---|
DTH | ||
Injection of an antigen intradermally and measurement of erythema and/or in duration after 48–72 hours | Easy to perform | Cutoff for positive response not standardized—subjective |
May correlate with T cell proliferative responses130 | Amount of antigen not standardized | |
May not be antigen specific131,132 | ||
May be a surrogate marker for better performance status/less immune suppression | ||
Peptide MHC tetramers | ||
Soluble, fluorescently labeled, MHC-peptide complex that binds to antigen-specific T cells | May correlate with cytotoxicity assays133 | Antigen must be known |
May correlate with T cell avidity for antigens134 | Tetramer positive cells may not kill target135 | |
Can be used to select antigen-specific T cell for further analysis136 | Only MHC I tetramers are available routinely | |
May bind both naïve and memory T cells137 | ||
Lymphoproliferative assay | ||
Purified T cells or PBMCs are stimulated with antigen in the presence of irradiated autologous APCs. After 72–100 hours, proliferation is measured and compared with the proliferative index of cells without antigen | Can be performed directly on peripheral blood samples | In vitro culture conditions can alter results |
May reflect the overall immune suppression state of patient | ||
High proliferation by a few cells or low levels of proliferation by a few cells would give a similar stimulation index | ||
ELISA/multiplex flow cytometric assay | ||
PBMCs are incubated with an antigen. Then the supernatant from the culture is harvested, and specific cytokines are detected | Easy to perform | Definition of positive results differs |
Is not based on individual cells | ||
Measures the ability of the cells to secrete cytokines and not necessarily the in vivo characteristics | ||
ELISPOT | ||
A microtiter plate is coated with a cytokine-specific antibody and then incubated with the T cells. Each spot represents a single cell secreting the cytokine of interest. Precursor frequency is determined by the total number of cells placed into the wells | Reliably detects the number of antigen-specific T cells138 | |
Can be rapidly read with computerized plate readers, making it suitable for a large-scale study | ||
Intracellular cytokine detection by multiparameter flow cytometry | ||
In vitro T cell stimulation followed by prevention of cytokine secretion, surface staining, fixation, permeabilization, and staining with antibody | Can evaluate multiple immune populations simultaneously | Cell viability is lost; so unable to perform subsequent functional assays |
Cytotoxicity assays | ||
Mix T cells or PBMCs with labeled antigen, expressing and measuring release of the target | Thought to be relevant marker for in vivo antitumor activity | Requires in vitro stimulation that may alter the activity from the in vivo state |
Because autologous tissue is difficult to obtain, other targets are used that may not reflect tumor biology | ||
Does not measure perforin, granzymes, or Fas-Fas-ligand cytotoxic killing—only direct cytotoxicity |
The best data available for defining an immune surrogate with clinical response come from infectious disease studies101–103 in which an increasing proportion of polyfunctional T cells, T cells that simultaneously secrete IFN-γ, tumor necrosis factor-2α, and IL-2 along with coordinated expression of CD107a as a marker for cytotoxicity, prospectively predict long-term nonprogressors in patients with human immunodeficiency and cytomegalovirus (CMV) infection.101,104,105 Increased numbers of polyfunctional antitumor T cells have also predicted improved antitumor efficacy, albeit only in animal models to date.106,107
In addition to polyfunctional T cell responses, there may be some hints in support of other immune surrogates in cancer. For example, Wheeler et al.108 showed a correlation between IFN-γ production and survival. In the phase III study that demonstrated the efficacy of sipuleucel-T (PA2024) (Provenge; Dendreon), the stimulation index of fresh T cells in response to antigen was ∼8-fold higher in the treatment group than in the controls.86 Of note, we conducted a similar analysis on the peptide-based vaccine targeting epidermal growth factor receptor vIII and used this same measure of immune response and were widely criticized during peer review.109 In addition to the lack of consistency and validation in T cell–based immunologic monitoring, immune monitoring has generally neglected other immune effectors such as monocytes, natural killer cells, and antibody-dependent cellular cytotoxicity, which may in fact be the mechanism of in vivo activity that should be more comprehensively evaluated in the context of these trials. In the interim, we should focus on validated surrogate immune markers from other fields and aggressively attempt to validate them in this field.
Toxicity and adverse events
The lack of significant toxicity, such as autoimmunity-associated, with immunotherapeutic approaches for GBM may indicate that insufficient immune responses are being generated for tumor eradication. In the case of melanoma immunotherapeutics, autoimmune responses have been shown to correlate with treatment response for both IL-2 and IFN-α.110,111 Although autoimmunity is not a perfect correlate of success in immunotherapy for melanoma, such autoimmune responses do indicate that robust immune responses against the target cell type can be obtained. A recent case of a patient with a brain metastasis from melanoma treated with cytotoxic T-lymphocyte antigen–4 antibodies demonstrated that when strong immune responses against brain antigens are unleashed, the immune response will probably have side effects.112 Similar adverse events were seen in the immunization trials against amyloid-β in the treatment of Alzheimer's disease in which 15% of patients developed severe encephalitis induced by T cells. Examination of the patients’ brains appeared to indicate that the inflammatory response was able to clear typical Alzheimer's neuropathology.113 Thus, if adverse events are an indication of strong effective immune stimulation, its absence from clinical immunotherapy trials could be of significance. However, enthusiasm for generating significant effector responses needs to be counterbalanced with the consideration that an expanding mass of inflammation within the relatively closed compartment of the CNS could result in herniation or fatal autoimmunity against CNS antigens.114 These adverse autoimmune events could also be reflective of the lack of tumor specificity. Thus, we recommend targeting TSA in the context of approaches that generate robust immune effector responses. However, this specificity can limit the durable response by the development of antigen negative tumor clones.115
Extrinsic Factors
We are in the midst of a transformation in the pharmaceutical industry, and it is one that may not be favorable for the development of novel therapeutics for primary brain tumors. Although the approval of TMZ demonstrated that drugs used for GBM can be profitable, this enthusiasm may not be sustained. According to Munos,116 pharmaceutical companies have on average developed only one new drug or biologic agent per year over the last 60 years. As a result, new drugs must be priced tremendously high and serve a large patient population to meet investors’ expectations, which means few, if any, drugs or biologics developed specifically for GBM will appear attractive. Moreover, large pharmaceutical companies, which have the capital to perform the required large and expensive phase III trials, are shifting their focus from early discovery and development to marketing. This is partially in response to demands from investors for nearer-term returns, but it leaves a significant void in early development that cannot be met by the current funding levels available to investigators from the National Institutes of Health. Even the cost of providing data for an initial investigational new drug application can mean an insurmountable financial burden (usually in the range of $1–2 million or more) for the investigator. This frequently does not even include manufacturing of the agent. Added to this is the cost for the initial clinical trials. Even for simple, off-the-shelf immunotherapeutics such as a peptide vaccine, the cost can be as high as $20 000 per patient/year in addition to the standard of care; thus, for a small and probably insufficiently powered clinical trial enrolling 25 patients, the cost to the investigator could be >$500 000.117 The price tag further escalates if the trial extends to other institutions because an extensive data and regulatory infrastructure needs to be put in place.
The increasing complexity of conducting clinical trials in the United States is further confounded by the increasing legal issue of indemnification. Multi-institutional clinical trials are advantageous because they rapidly enroll patients and may reduce institutional bias; however, negotiating participating institution indemnification can result in unexpected delays and further escalate clinical trial costs. Increasingly, as a result of this and other financial pressures, pharmaceutical companies are conducting clinical trials outside the United States.118 In fact, in just under 10 years (1997–2007), the percentage of clinical trials registered with the FDA to be conducted in the United States dropped from 86% to 57%, whereas sites in places such as India and China rose from 5% to 29%.119 As a result, very creative strategies will be needed to translate any immunotherapeutic for GBM beyond the dubious single-institution early-phase trial and to demonstrate sufficient efficacy to attract the interest of companies with sufficient resources to bring these agents to market. These extrinsic factors pose a real threat to the emergence of a standard-of-care immunotherapy for GBM and will need to be addressed creatively.
Conclusions and Recommendations
Although the benefits of immunotherapy are becoming evident in other fields, its use in neuro-oncology remains limited. To advance these promising approaches toward a standard of care will require that lessons learned from basic science investigations in neuro-immunology and immunotherapy in other fields be applied creatively and cost-effectively. A number of conclusions and recommendations can be derived from our review.
Immune responses exert therapeutic effects within the CNS; therefore, immunotherapy is a viable approach to CNS malignancies.
Although tumors within the CNS are not completely protected from an immune attack, sufficiently potent immune responses need to be generated to overcome profound immunosuppression, or the immunosuppression has to be minimized by tumor resection or with agents that target tumor-mediated immunosuppression globally or at key molecular hubs. Successful approaches will probably incorporate both.
Clinical trial design should be carefully considered. Traditional paradigms may not be informative. An immune response endpoint may not yield meaningful results. Given the recent shifting in GBM patient survival time, randomization should be strongly considered even during early testing. Furthermore, the selection criteria during early phases of clinical trial testing should be the same as in the final registration clinical trial. Immunotherapeutic prognostic markers need to be identified and accounted for as secondary endpoints.
Immunologic surrogates that predict the efficacy of immunotherapeutic approaches in cancer are not currently available but are desperately needed. Clues as to which responses are important may come from existing studies or from infectious disease investigations and may not be as expected. Effective antitumor immune responses may require coordinated actions among several components of the immune system, all of which may need to be monitored. Immune monitoring results will become useful only when they are standardized and prospectively validated.
New and creative development and marketing paradigms will definitely be needed if we are to achieve translation of immunotherapeutics for brain tumors into widely used therapeutics.
Funding
Funding support came from the following grants: NIH –(5RO1-CA135272-02, 3RO1-CA135272-02S1, 5P50-CA108786-05, 5P50-CA127001-03, 1R25-NS065731-01, 5P50-NS020023-27, 5R21-CA132891-02, 3R21CA132891-02S1, R01-CA097222, R01-CA097611, P50CA108786, R01-CA1208113, R43 AI 77225-01), Golfers Against Cancer, National Brain Tumor Foundation Translational Research Grant, Neurosurgery Research and Education Foundation, Shawn Hansen/American Brain Tumor Association, Adam Sliger Research Fund, Goodwin Fund for Clinical Trials Grant, National Brain Tumor Society, Dr Marnie Rose Foundation, Anthony Bullock III Foundation, PBTFUS, The Brain Tumor Society, Congress of Neurological Surgeons, Southeastern Brain Tumor Foundation, Accelerate Brain Cancer Cure/American Brain Tumor Association, AANS/CNS, and the Chandran Research Award.
Acknowledgments
We thank Jim Herndon II, PhD, for statistical assistance. We thank Audria Patrick and David Wildrick, PhD, for editorial assistance.
Conflicts of interest statement. The authors and their respective institutions of The University of Texas MD Anderson Cancer Center and Duke University have received consulting fees, stock options, and licensing fees from Celldex Therapeutics and Pfizer.
References
- 1.Medawar PB. Immunity to homologous grafted skin. III. The fate of skin homografts transplanted to the brain, to subcutaneous tissue, and to the anterior chamber of the eye. Br J Exp Pathol. 1948;29:58–69. [PMC free article] [PubMed] [Google Scholar]
- 2.Fabry Z, Raine CS, Hart MN. Nervous tissue as an immune compartment: the dialect of the immune response in the CNS. Immunol Today. 1994;15:218–224. doi: 10.1016/0167-5699(94)90247-X. [DOI] [PubMed] [Google Scholar]
- 3.Parney IF, Hao C, Petruk KC. Glioma immunology and immunotherapy. Neurosurgery. 2000;46:778–791. doi: 10.1097/00006123-200004000-00002. discussion 791–792. [DOI] [PubMed] [Google Scholar]
- 4.Cserr HF, Knopf PM. Cervical lymphatics, the blood-brain barrier and the immunoreactivity of the brain: a new view. Immunol Today. 1992;13:507–512. doi: 10.1016/0167-5699(92)90027-5. [DOI] [PubMed] [Google Scholar]
- 5.Cserr HF, Harling-Berg CJ, Knopf PM. Drainage of brain extracellular fluid into blood and deep cervical lymph and its immunological significance. Brain Pathol. 1992;2:269–276. doi: 10.1111/j.1750-3639.1992.tb00703.x. [DOI] [PubMed] [Google Scholar]
- 6.Harling-Berg CJ, Park TJ, Knopf PM. Role of the cervical lymphatics in the Th2-type hierarchy of CNS immune regulation. J Neuroimmunol. 1999;101:111–127. doi: 10.1016/s0165-5728(99)00130-7. [DOI] [PubMed] [Google Scholar]
- 7.Weller RO, Engelhardt B, Phillips MJ. Lymphocyte targeting of the central nervous system: a review of afferent and efferent CNS-immune pathways. Brain Pathol. 1996;6:275–288. doi: 10.1111/j.1750-3639.1996.tb00855.x. [DOI] [PubMed] [Google Scholar]
- 8.Harling-Berg C, Knopf PM, Merriam J, Cserr HF. Role of cervical lymph nodes in the systemic humoral immune response to human serum albumin microinfused into rat cerebrospinal fluid. J Neuroimmunol. 1989;25:185–193. doi: 10.1016/0165-5728(89)90136-7. [DOI] [PubMed] [Google Scholar]
- 9.Harling-Berg CJ, Hallett JJ, Park JT, Knopf PM. Hierarchy of immune responses to antigen in the normal brain. Curr Top Microbiol Immunol. 2002;265:1–22. doi: 10.1007/978-3-662-09525-6_1. [DOI] [PubMed] [Google Scholar]
- 10.Mosmann TR, Coffman RL. TH1 and TH2 cells: different patterns of lymphokine secretion lead to different functional properties. Annu Rev Immunol. 1989;7:145–173. doi: 10.1146/annurev.iy.07.040189.001045. [DOI] [PubMed] [Google Scholar]
- 11.Bullard DE, Gillespie GY, Mahaley MS, Bigner DD. Immunobiology of human gliomas. Semin Oncol. 1986;13:94–109. [PubMed] [Google Scholar]
- 12.Owens T, Renno T, Taupin V, Krakowski M. Inflammatory cytokines in the brain: does the CNS shape immune responses. Immunol Today. 1994;15:566–571. doi: 10.1016/0167-5699(94)90218-6. [DOI] [PubMed] [Google Scholar]
- 13.Stevens A, Kloter I, Roggendorf W. Inflammatory infiltrates and natural killer cell presence in human brain tumors. Cancer. 1988;61:738–743. doi: 10.1002/1097-0142(19880215)61:4<738::aid-cncr2820610417>3.0.co;2-e. [DOI] [PubMed] [Google Scholar]
- 14.Hickey WF, Hsu BL, Kimura H. T-lymphocyte entry into the central nervous system. J Neurosci Res. 1991;28:254–260. doi: 10.1002/jnr.490280213. [DOI] [PubMed] [Google Scholar]
- 15.Goldmann J, Kwidzinski E, Brandt C, Mahlo J, Richter D, Bechmann I. T cells traffic from brain to cervical lymph nodes via the cribroid plate and the nasal mucosa. J Leukoc Biol. 2006;80:797–801. doi: 10.1189/jlb.0306176. [DOI] [PubMed] [Google Scholar]
- 16.Masson F, Calzascia T, Di Berardino-Besson W, de Tribolet N, Dietrich PY, Walker PR. Brain microenvironment promotes the final functional maturation of tumor-specific effector CD8+ T cells. J Immunol. 2007;179:845–853. doi: 10.4049/jimmunol.179.2.845. [DOI] [PubMed] [Google Scholar]
- 17.Aloisi F, Ria F, Adorini L. Regulation of T-cell responses by CNS antigen-presenting cells: different roles for microglia and astrocytes. Immunol Today. 2000;21:141–147. doi: 10.1016/s0167-5699(99)01512-1. [DOI] [PubMed] [Google Scholar]
- 18.Irani DN, Lin KI, Griffin DE. Regulation of brain-derived T cells during acute central nervous system inflammation. J Immunol. 1997;158:2318–2326. [PubMed] [Google Scholar]
- 19.Gehrmann J, Banati RB, Wiessner C, Hossmann KA, Kreutzberg GW. Reactive microglia in cerebral ischaemia: an early mediator of tissue damage? Neuropathol Appl Neurobiol. 1995;21:277–289. doi: 10.1111/j.1365-2990.1995.tb01062.x. [DOI] [PubMed] [Google Scholar]
- 20.Serot JM, Foliguet B, Bene MC, Faure GC. Ultrastructural and immunohistological evidence for dendritic-like cells within human choroid plexus epithelium. Neuroreport. 1997;8:1995–1998. doi: 10.1097/00001756-199705260-00039. [DOI] [PubMed] [Google Scholar]
- 21.McMenamin PG, Forrester JV. Dendritic Cells: Biology and Clinical Applications. New York: Academic Press; 1999. Dendritic cells in the central nervous system and eye and their associated supporting tissues; pp. 205–248. [Google Scholar]
- 22.Hickey WF, Kimura H. Perivascular microglial cells of the CNS are bone marrow-derived and present antigen in vivo. Science. 1988;239:290–292. doi: 10.1126/science.3276004. [DOI] [PubMed] [Google Scholar]
- 23.Lowe J, MacLennan KA, Powe DG, Pound JD, Palmer JB. Microglial cells in human brain have phenotypic characteristics related to possible function as dendritic antigen presenting cells. J Pathol. 1989;159:143–149. doi: 10.1002/path.1711590209. [DOI] [PubMed] [Google Scholar]
- 24.Ulvestad E, Williams K, Bjerkvig R, Tiekotter K, Antel J, Matre R. Human microglial cells have phenotypic and functional characteristics in common with both macrophages and dendritic antigen-presenting cells. J Leukoc Biol. 1994;56:732–740. doi: 10.1002/jlb.56.6.732. [DOI] [PubMed] [Google Scholar]
- 25.Gehrmann J, Banati RB, Kreutzberg GW. Microglia in the immune surveillance of the brain: human microglia constitutively express HLA-DR molecules. J Neuroimmunol. 1993;48:189–198. doi: 10.1016/0165-5728(93)90191-z. [DOI] [PubMed] [Google Scholar]
- 26.Williams K, Jr, Ulvestad E, Cragg L, Blain M, Antel JP. Induction of primary T cell responses by human glial cells. J Neurosci Res. 1993;36:382–390. doi: 10.1002/jnr.490360404. [DOI] [PubMed] [Google Scholar]
- 27.Furr M. Antigen-specific antibodies in cerebrospinal fluid after intramuscular injection of ovalbumin in horses. J Vet Intern Med. 2002;16:588–592. doi: 10.1892/0891-6640(2002)016<0588:aaicfa>2.3.co;2. [DOI] [PubMed] [Google Scholar]
- 28.Gigliotti F, Lee D, Insel RA, Scheld WM. IgG penetration into the cerebrospinal fluid in a rabbit model of meningitis. J Infect Dis. 1987;156:394–398. doi: 10.1093/infdis/156.2.394. [DOI] [PubMed] [Google Scholar]
- 29.Pestalozzi BC, Brignoli S. Trastuzumab in CSF. J Clin Oncol. 2000;18:2349–2351. doi: 10.1200/JCO.2000.18.11.2349. [DOI] [PubMed] [Google Scholar]
- 30.Stemmler HJ, Schmitt M, Willems A, Bernhard H, Harbeck N, Heinemann V. Ratio of trastuzumab levels in serum and cerebrospinal fluid is altered in HER2-positive breast cancer patients with brain metastases and impairment of blood-brain barrier. Anticancer Drugs. 2007;18:23–28. doi: 10.1097/01.cad.0000236313.50833.ee. [DOI] [PubMed] [Google Scholar]
- 31.Kallioniemi OP, Holli K, Visakorpi T, Koivula T, Helin HH, Isola JJ. Association of c-erbB-2 protein over-expression with high rate of cell proliferation, increased risk of visceral metastasis and poor long-term survival in breast cancer. Int J Cancer. 1991;49:650–655. doi: 10.1002/ijc.2910490504. [DOI] [PubMed] [Google Scholar]
- 32.Pestalozzi BC. Brain metastases and subtypes of breast cancer. Ann Oncol. 2009;20:803–805. doi: 10.1093/annonc/mdp246. [DOI] [PubMed] [Google Scholar]
- 33.de Vries E. 11th International Conference; St. Gallen, Switzerland, 2009. [Google Scholar]
- 34.Dey M, Hussain SF, Heimberger AB. The role of glioma microenvironment in immune modulation: potential targets for intervention. Lett Drug Des Discov. 2006;3:443–451. [Google Scholar]
- 35.Calzascia T, Masson F, Di Berardino-Besson W, et al. Homing phenotypes of tumor-specific CD8 T cells are predetermined at the tumor site by crosspresenting APCs. Immunity. 2005;22:175–184. doi: 10.1016/j.immuni.2004.12.008. [DOI] [PubMed] [Google Scholar]
- 36.Hussain SF, Yang D, Suki D, Aldape K, Grimm E, Heimberger AB. The role of human glioma-infiltrating microglia/macrophages in mediating antitumor immune responses. Neurooncology. 2006;8:261–279. doi: 10.1215/15228517-2006-008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Platten M, Wick W, Weller M. Malignant glioma biology: role for TGF-beta in growth, motility, angiogenesis, and immune escape. Microsc Res Tech. 2001;52:401–410. doi: 10.1002/1097-0029(20010215)52:4<401::AID-JEMT1025>3.0.CO;2-C. [DOI] [PubMed] [Google Scholar]
- 38.Roszman T, Elliott L, Brooks W. Modulation of T-cell function by gliomas [Review] Immunol Today. 1991;12:370–374. doi: 10.1016/0167-5699(91)90068-5. [DOI] [PubMed] [Google Scholar]
- 39.Tada M, Suzuki K, Yamakawa Y, et al. Human glioblastoma cells produce 77 amino acid interleukin-8 (IL- 8(77)) J Neurooncol. 1993;16:25–34. doi: 10.1007/BF01324831. [DOI] [PubMed] [Google Scholar]
- 40.Gabrilovich D, Ishida T, Oyama T, et al. Vascular endothelial growth factor inhibits the development of dendritic cells and dramatically affects the differentiation of multiple hematopoietic lineages in vivo. Blood. 1998;92:4150–4166. [PubMed] [Google Scholar]
- 41.Tirapu I, Huarte E, Guiducci C, et al. Low surface expression of B7–1 (CD80) is an immunoescape mechanism of colon carcinoma. Cancer Res. 2006;66:2442–2450. doi: 10.1158/0008-5472.CAN-05-1681. [DOI] [PubMed] [Google Scholar]
- 42.Voigt H, Schrama D, Eggert AO, et al. CD28-mediated costimulation impacts on the differentiation of DC vaccination-induced T cell responses. Clin Exp Immunol. 2006;143:93–102. doi: 10.1111/j.1365-2249.2005.02972.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Yi-qun Z, Lorre K, de Boer M, Ceuppens JL. B7-blocking agents, alone or in combination with cyclosporin A, induce antigen-specific anergy of human memory T cells. J Immunol. 1997;158:4734–4740. [PubMed] [Google Scholar]
- 44.Matyszak MK, Denis-Donini S, Citterio S, Longhi R, Granucci F, Ricciardi-Castagnoli P. Microglia induce myelin basic protein-specific T cell anergy or T cell activation, according to their state of activation. Eur J Immunol. 1999;29:3063–3076. doi: 10.1002/(SICI)1521-4141(199910)29:10<3063::AID-IMMU3063>3.0.CO;2-G. [DOI] [PubMed] [Google Scholar]
- 45.Parsa AT, Waldron JS, Panner A, et al. Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma. Nat Med. 2007;13:84–88. doi: 10.1038/nm1517. [DOI] [PubMed] [Google Scholar]
- 46.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. 2005;28:582–592. doi: 10.1097/01.cji.0000175468.19742.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Berd D, Mastrangelo MJ. Effect of low dose cyclophosphamide on the immune system of cancer patients: reduction of T-suppressor function without depletion of the CD8+ subset. Cancer Res. 1987;47:3317–3321. [PubMed] [Google Scholar]
- 48.Foss F. Clinical experience with denileukin diftitox (ONTAK) Semin Oncol. 2006;33:11–16. doi: 10.1053/j.seminoncol.2005.12.017. [DOI] [PubMed] [Google Scholar]
- 49.Sakaguchi S. Naturally arising CD4+ regulatory T cells for immunologic self-tolerance and negative control of immune responses. Annu Rev Immunol. 2004;22:531–562. doi: 10.1146/annurev.immunol.21.120601.141122. [DOI] [PubMed] [Google Scholar]
- 50.Su YB, Sohn S, Krown SE, et al. Selective CD4+ lymphopenia in melanoma patients treated with temozolomide: a toxicity with therapeutic implications. J Clin Oncol. 2004;22:610–616. doi: 10.1200/JCO.2004.07.060. [DOI] [PubMed] [Google Scholar]
- 51.Curiel TJ, Coukos G, Zou L, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med. 2004;10:942–949. doi: 10.1038/nm1093. [DOI] [PubMed] [Google Scholar]
- 52.El Andaloussi A, Lesniak MS. CD4+ CD25+ FoxP3+ T-cell infiltration and heme oxygenase-1 expression correlate with tumor grade in human gliomas. J Neurooncol. 2007;83:145–152. doi: 10.1007/s11060-006-9314-y. [DOI] [PubMed] [Google Scholar]
- 53.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. 2006;66:3294–3302. doi: 10.1158/0008-5472.CAN-05-3773. [DOI] [PubMed] [Google Scholar]
- 54.Heimberger AB, Reina-Ortiz C, Yang DS, et al. Incidence and prognostic impact of FoxP3+ regulatory T cells in human gliomas. Clin Cancer Res. 2008;14:5166–5172. doi: 10.1158/1078-0432.CCR-08-0320. [DOI] [PubMed] [Google Scholar]
- 55.Umemura N, Saio M, Suwa T, et al. Tumor-infiltrating myeloid-derived suppressor cells are pleiotropic-inflamed monocytes/macrophages that bear M1- and M2-type characteristics. J Leukoc Biol. 2008;83:1136–1144. doi: 10.1189/jlb.0907611. [DOI] [PubMed] [Google Scholar]
- 56.Mantovani A, Sica A, Allavena P, Garlanda C, Locati M. Tumor-associated macrophages and the related myeloid-derived suppressor cells as a paradigm of the diversity of macrophage activation. Hum Immunol. 2009;70:325–330. doi: 10.1016/j.humimm.2009.02.008. [DOI] [PubMed] [Google Scholar]
- 57.Pollard JW. Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer. 2004;4:71–78. doi: 10.1038/nrc1256. [DOI] [PubMed] [Google Scholar]
- 58.Di Tomaso T, Mazzoleni S, Wang E, et al. Immunobiological characterization of cancer stem cells isolated from glioblastoma patients. Clin Cancer Res. 2010;16:800–813. doi: 10.1158/1078-0432.CCR-09-2730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Schatton T, Schutte U, Frank NY, et al. Modulation of T-cell activation by malignant melanoma initiating cells. Cancer Res. 2010;70:697–708. doi: 10.1158/0008-5472.CAN-09-1592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Wei J, Bar J, Kong L-Y, et al. Glioblastoma cancer-initiating cells inhibit T cell proliferation and effector responses by the STAT3 pathway. Mol Cancer Ther. 2010;9:67–78. doi: 10.1158/1535-7163.MCT-09-0734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Wei J, Barr J, Kong L-Y, et al. Glioma associated cancer-initiating cells induce immune suppression. Clin Cancer Res. 2010;16:461–473. doi: 10.1158/1078-0432.CCR-09-1983. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 62.Brown CE, Starr R, Martinez C, et al. Recognition and killing of brain tumor stem-like initiating cells by CD8+ cytolytic T cells. Cancer Res. 2009;69:8886–8893. doi: 10.1158/0008-5472.CAN-09-2687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Pellegatta S, Finocchiaro G. Dendritic cell vaccines for cancer stem cells. Methods Mol Biol. 2009;568:233–247. doi: 10.1007/978-1-59745-280-9_15. [DOI] [PubMed] [Google Scholar]
- 64.Xu Q, Liu G, Yuan X, et al. Antigen-specific T-cell response from dendritic cell vaccination using cancer stem-like cell-associated antigens. Stem Cells. 2009;27:1734–1740. doi: 10.1002/stem.102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Lang R, Patel D, Morris JJ, Rutschman RL, Murray PJ. Shaping gene expression in activated and resting primary macrophages by IL-10. J Immunol. 2002;169:2253–2263. doi: 10.4049/jimmunol.169.5.2253. [DOI] [PubMed] [Google Scholar]
- 66.O'Farrell AM, Liu Y, Moore KW, Mui AL. IL-10 inhibits macrophage activation and proliferation by distinct signaling mechanisms: evidence for Stat3-dependent and independent pathways. EMBO J. 1998;17:1006–1018. doi: 10.1093/emboj/17.4.1006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Takeda K, Clausen BE, Kaisho T, et al. Enhanced Th1 activity and development of chronic enterocolitis in mice devoid of Stat3 in macrophages and neutrophils. Immunity. 1999;10:39–49. doi: 10.1016/s1074-7613(00)80005-9. [DOI] [PubMed] [Google Scholar]
- 68.Lin T, Bost K. STAT3 activation in macrophages following infection with Salmonella. Biochem Biophys Res Commun. 2004;321:828–834. doi: 10.1016/j.bbrc.2004.07.039. [DOI] [PubMed] [Google Scholar]
- 69.Kortylewski M, Kujawski M, Wang T, et al. Inhibiting Stat3 signaling in the hematopoietic system elicits multicomponent antitumor immunity. Nat Med. 2005;11:1314–1321. doi: 10.1038/nm1325. [DOI] [PubMed] [Google Scholar]
- 70.Wu A, Wei J, Kong LY, et al. Glioma cancer stem cells induce immunosuppressive macrophages/microglia. Neuro Oncol. 2010;12:1113–1125. doi: 10.1093/neuonc/noq082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Kong L-K, Wei J, Sharma AK, et al. A novel phosphorylated STAT3 inhibitor enhances T cell cytotoxicity against melanoma through inhibition of regulatory T cells. Cancer Immunol Immunother. 2008;58:1023–1032. doi: 10.1007/s00262-008-0618-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Kong LY, Abou-Ghazal MK, Wei J, et al. A novel inhibitor of signal transducers and activators of transcription 3 activation is efficacious against established central nervous system melanoma and inhibits regulatory T cells. Clin Cancer Res. 2008;14:5759–5768. doi: 10.1158/1078-0432.CCR-08-0377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Zorn E, Nelson EA, Mohseni M, et al. IL-2 regulates FOXP3 expression in human CD4+CD25+ regulatory T cells through a STAT-dependent mechanism and induces the expansion of these cells in vivo. Blood. 2006;108:1571–1579. doi: 10.1182/blood-2006-02-004747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Zhang JG, Kruse CA, Driggers L, et al. Tumor antigen precursor protein profiles of adult and pediatric brain tumors identifies potential targets for immunotherapy. J Neuro-Oncol. 2008;88:65–76. doi: 10.1007/s11060-008-9534-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.He XS, Holmes TH, Zhang C, et al. Cellular immune responses in children and adults receiving inactivated or live attenuated influenza vaccines. J Virol. 2006;80:11756–11766. doi: 10.1128/JVI.01460-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Postel-Vinay S, Arkenau HT, Olmos D, et al. Clinical benefit in Phase-I trials of novel molecularly targeted agents: does dose matter. Br J Cancer. 2009;100:1373–1378. doi: 10.1038/sj.bjc.6605030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Suntharalingam G, Perry MR, Ward S, et al. Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. N Engl J Med. 2006;355:1018–1028. doi: 10.1056/NEJMoa063842. [DOI] [PubMed] [Google Scholar]
- 78.Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352:987–996. doi: 10.1056/NEJMoa043330. [DOI] [PubMed] [Google Scholar]
- 79.Rhee DJ, Kong DS, Kim WS, et al. Efficacy of temozolomide as adjuvant chemotherapy after postsurgical radiotherapy alone for glioblastomas. Clin Neurol Neurosurg. 2009;111:748–751. doi: 10.1016/j.clineuro.2009.07.014. [DOI] [PubMed] [Google Scholar]
- 80.Grossman SA, Ye X, Piantadosi S, et al. Survival of patients with newly diagnosed glioblastoma treated with radiation and temozolomide in research studies in the United States. Clin Cancer Res. 2010;16:2443–2449. doi: 10.1158/1078-0432.CCR-09-3106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Shahar T, Nossek E, Blumenthal DT, et al. Trial to survive: the impact of enrollment in clinical trials on survival of patients with glioblastoma. Neurosurgery. 2009;65:425. doi: 10.1016/j.jocn.2012.04.005. [DOI] [PubMed] [Google Scholar]
- 82.Kirkwood JM, Manola J, Ibrahim J, Sondak V, Ernstoff MS, Rao U. A pooled analysis of eastern cooperative oncology group and intergroup trials of adjuvant high-dose interferon for melanoma. Clin Cancer Res. 2004;10:1670–1677. doi: 10.1158/1078-0432.ccr-1103-3. [DOI] [PubMed] [Google Scholar]
- 83.Atkins MB, Kunkel L, Sznol M, Rosenberg SA. High-dose recombinant interleukin-2 therapy in patients with metastatic melanoma: long-term survival update. Cancer J Sci Am. 2000;6(suppl 1):S11–S14. [PubMed] [Google Scholar]
- 84.Atkins MB, Lotze MT, Dutcher JP, et al. High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993. J Clin Oncol. 1999;17:2105–2116. doi: 10.1200/JCO.1999.17.7.2105. [DOI] [PubMed] [Google Scholar]
- 85.Rosenberg SA, Yang JC, Topalian SL, et al. Treatment of 283 consecutive patients with metastatic melanoma or renal cell cancer using high-dose bolus interleukin 2. JAMA. 1994;271:907–913. [PubMed] [Google Scholar]
- 86.Small EJ, Schellhammer PF, Higano CS, et al. Placebo-controlled phase III trial of immunologic therapy with sipuleucel-T (APC8015) in patients with metastatic, asymptomatic hormone refractory prostate cancer. J Clin Oncol. 2006;24:3089–3094. doi: 10.1200/JCO.2005.04.5252. [DOI] [PubMed] [Google Scholar]
- 87.Hodi FS, O'Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–723. doi: 10.1056/NEJMoa1003466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.El Andaloussi A, Lesniak MS. An increase in CD4+CD25+FOXP3+ regulatory T cells in tumor-infiltrating lymphocytes of human glioblastoma multiforme. Neurooncology. 2006;8:234–243. doi: 10.1215/15228517-2006-006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.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. 2006;12:4294–4305. doi: 10.1158/1078-0432.CCR-06-0053. [DOI] [PubMed] [Google Scholar]
- 90.Carro MS, Lim WK, Alvarez MJ, et al. The transcriptional network for mesenchymal transformation of brain tumours. Nature. 2010;463:318–325. doi: 10.1038/nature08712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Freije WA, Castro-Vargas FE, Fang Z, et al. Gene expression profiling of gliomas strongly predicts survival. Cancer Res. 2004;64:6503–6510. doi: 10.1158/0008-5472.CAN-04-0452. [DOI] [PubMed] [Google Scholar]
- 92.Murat A, Migliavacca E, Gorlia T, et al. Stem cell-related “self-renewal” signature and high epidermal growth factor receptor expression associated with resistance to concomitant chemoradiotherapy in glioblastoma. J Clin Oncol. 2008;26:3015–3024. doi: 10.1200/JCO.2007.15.7164. [DOI] [PubMed] [Google Scholar]
- 93.Murat A, Migliavacca E, Hussain SF, et al. Modulation of angiogenic and inflammatory response in glioblastoma by hypoxia. PLoS One. 2009;4:e5947. doi: 10.1371/journal.pone.0005947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Hoover HC, Jr, Surdyke M, Dangel RB, Peters LC, Hanna MG., Jr Delayed cutaneous hypersensitivity to autologous tumor cells in colorectal cancer patients immunized with an autologous tumor cell: Bacillus Calmette-Guerin vaccine. Cancer Res. 1984;44:1671–1676. [PubMed] [Google Scholar]
- 95.Khleif SN, Abrams SI, Hamilton JM, et al. A phase I vaccine trial with peptides reflecting ras oncogene mutations of solid tumors. J Immunother. 1999;22:155–165. doi: 10.1097/00002371-199903000-00007. [DOI] [PubMed] [Google Scholar]
- 96.Maraveyas A, Baban B, Kennard D, et al. Possible improved survival of patients with stage IV AJCC melanoma receiving SRL 172 immunotherapy: correlation with induction of increased levels of intracellular interleukin-2 in peripheral blood lymphocytes. Ann Oncol. 1999;10:817–824. doi: 10.1023/a:1008307821189. [DOI] [PubMed] [Google Scholar]
- 97.Marchand M, van Baren N, Weynants P, et al. Tumor regressions observed in patients with metastatic melanoma treated with an antigenic peptide encoded by gene MAGE-3 and presented by HLA-A1. Int J Cancer. 1999;80:219–230. doi: 10.1002/(sici)1097-0215(19990118)80:2<219::aid-ijc10>3.0.co;2-s. [DOI] [PubMed] [Google Scholar]
- 98.Nestle FO, Alijagic S, Gilliet M, et al. Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nat Med. 1998;4:328–332. doi: 10.1038/nm0398-328. [DOI] [PubMed] [Google Scholar]
- 99.Reynolds SR, Oratz R, Shapiro RL, et al. Stimulation of CD8+ T cell responses to MAGE-3 and Melan A/MART-1 by immunization to a polyvalent melanoma vaccine. Int J Cancer. 1997;72:972–976. doi: 10.1002/(sici)1097-0215(19970917)72:6<972::aid-ijc9>3.0.co;2-m. [DOI] [PubMed] [Google Scholar]
- 100.Schreiber S, Kampgen E, Wagner E, et al. Immunotherapy of metastatic malignant melanoma by a vaccine consisting of autologous interleukin 2-transfected cancer cells: outcome of a phase I study. Hum Gene Ther. 1999;10:983–993. doi: 10.1089/10430349950018382. [DOI] [PubMed] [Google Scholar]
- 101.Betts MR, Nason MC, West SM, et al. HIV nonprogressors preferentially maintain highly functional HIV-specific CD8+ T cells. Blood. 2006;107:4781–4789. doi: 10.1182/blood-2005-12-4818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Lindenstrom T, Agger EM, Korsholm KS, et al. Tuberculosis subunit vaccination provides long-term protective immunity characterized by multifunctional CD4 memory T cells. J Immunol. 2009;182:8047–8055. doi: 10.4049/jimmunol.0801592. [DOI] [PubMed] [Google Scholar]
- 103.Seder RA, Darrah PA, Roederer M. T-cell quality in memory and protection: implications for vaccine design. Nat Rev Immunol. 2008;8:247–258. doi: 10.1038/nri2274. [DOI] [PubMed] [Google Scholar]
- 104.Betts MR, Exley B, Price DA, et al. Characterization of functional and phenotypic changes in anti-Gag vaccine-induced T cell responses and their role in protection after HIV-1 infection. Proc Natl Acad Sci USA. 2005;102:4512–4517. doi: 10.1073/pnas.0408773102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Betts MR, Gray CM, Cox JH, Ferrari G. Antigen-specific T-cell-mediated immunity after HIV-1 infection: implications for vaccine control of HIV development. Expert Rev Vaccines. 2006;5:505–516. doi: 10.1586/14760584.5.4.505. [DOI] [PubMed] [Google Scholar]
- 106.Rizzuto GA, Merghoub T, Hirschhorn-Cymerman D, et al. Self-antigen-specific CD8+ T cell precursor frequency determines the quality of the antitumor immune response. J Exp Med. 2009;206:849–866. doi: 10.1084/jem.20081382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Yuan J, Gnjatic S, Li H, et al. CTLA-4 blockade enhances polyfunctional NY-ESO-1 specific T cell responses in metastatic melanoma patients with clinical benefit. Proc Natl Acad Sci USA. 2008;105:20410–20415. doi: 10.1073/pnas.0810114105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Wheeler CJ, Black KL, Liu G, et al. Vaccination elicits correlated immune and clinical responses in glioblastoma multiforme patients. Cancer Res. 2008;68:5955–5964. doi: 10.1158/0008-5472.CAN-07-5973. [DOI] [PubMed] [Google Scholar]
- 109.Sampson JH, Archer GE, Mitchell DA, et al. An epidermal growth factor receptor variant III-targeted vaccine is safe and immunogenic in patients with glioblastoma multiforme. Mol Cancer Ther. 2009;8:2773–2779. doi: 10.1158/1535-7163.MCT-09-0124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Boasberg PD, Hoon DS, Piro LD, et al. Enhanced survival associated with vitiligo expression during maintenance biotherapy for metastatic melanoma. J Invest Dermatol. 2006;126:2658–2663. doi: 10.1038/sj.jid.5700545. [DOI] [PubMed] [Google Scholar]
- 111.Gogas H, Ioannovich J, Dafni U, et al. Prognostic significance of autoimmunity during treatment of melanoma with interferon. N Engl J Med. 2006;354:709–718. doi: 10.1056/NEJMoa053007. [DOI] [PubMed] [Google Scholar]
- 112.Kaehler KC, Egberts F, Lorigan P, Hauschild A. Anti-CTLA-4 therapy-related autoimmune hypophysitis in a melanoma patient. Melanoma Res. 2009;19:333–334. doi: 10.1097/CMR.0b013e32832e0bff. [DOI] [PubMed] [Google Scholar]
- 113.Orgogozo JM, Gilman S, Dartigues JF, et al. Subacute meningoencephalitis in a subset of patients with AD after Abeta42 immunization. Neurology. 2003;61:46–54. doi: 10.1212/01.wnl.0000073623.84147.a8. [DOI] [PubMed] [Google Scholar]
- 114.Wikstrand CJ, Bigner DD. Hyperimmunization of non-human primates with BCG-CW and cultured human glioma-derived cells. Production of reactive antisera and absence of EAE induction. J Neuroimmunol. 1981;1:249–260. doi: 10.1016/0165-5728(81)90029-1. [DOI] [PubMed] [Google Scholar]
- 115.Heimberger AB, Crotty LE, Archer GE, et al. Epidermal growth factor receptor VIII peptide vaccination is efficacious against established intracerebral tumors. Clin Cancer Res. 2003;9:4247–4254. [PubMed] [Google Scholar]
- 116.Munos B. Lessons from 60 years of pharmaceutical innovation. Nat Rev Drug Discov. 2009;8:959–968. doi: 10.1038/nrd2961. [DOI] [PubMed] [Google Scholar]
- 117.PRNewswire, vol. 2010. 2006 [Google Scholar]
- 118.Nundy S, Gulhati CM. A new colonialism?—conducting clinical trials in India. N Engl J Med. 2005;352:1633–1636. doi: 10.1056/NEJMp048361. [DOI] [PubMed] [Google Scholar]
- 119.Outsourcing clinical trials: is it ethical to take drug studies abroad. O'Reilly KB. 2009. [cited 2010 February 18]; Available from: http://www.ama-assn.org/amednews/2009/09/07/prsa0907.htm .
- 120.Sampson JH, Heimberger AB, Archer G, et al. Immunologic escape after prolonged progression-free survival with epidermal growth factor receptor variant III peptide vaccination in patients with newly diagnosed glioblastoma. J Clin Oncol. 2010;28:4722–4729. doi: 10.1200/JCO.2010.28.6963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Sampson JH, Archer GE, Bigner DD, et al. Effect of EGFRvIII-targeted vaccine (CDX-110) on immune response and TTP when given with simultaneous standard and continuous temozolomide in patients with GBM. J Clin Oncol (Meeting Abstracts) 2008;26:2011. [Google Scholar]
- 122.Yajima N, Yamanaka R, Mine T, et al. Immunologic evaluation of personalized peptide vaccination for patients with advanced malignant glioma. Clin Cancer Res. 2005;11:5900–5911. doi: 10.1158/1078-0432.CCR-05-0559. [DOI] [PubMed] [Google Scholar]
- 123.Mitchell DA, Archer G, Bigner D, et al. RNA-loaded dendritic cells targeting cytomegalovirus in patients with malignant glioma. Neurooncology. 2007;9:509. [Google Scholar]
- 124.De Vleeschouwer S, Fieuws S, Rutkowski S, et al. Postoperative adjuvant dendritic cell-based immunotherapy in patients with relapsed glioblastoma multiforme. Clin Cancer Res. 2008;14:3098–3104. doi: 10.1158/1078-0432.CCR-07-4875. [DOI] [PubMed] [Google Scholar]
- 125.Yu J, Liu G, Ying H, Yong W, Black K, Wheeler CJ. Vaccination with tumor lysate-pulsed dendritic cells elicits antigen-specific, cytotoxic T-cells in patients with malignant glioma. Cancer Res. 2004;64:4973–4979. doi: 10.1158/0008-5472.CAN-03-3505. [DOI] [PubMed] [Google Scholar]
- 126.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. 2005;11:5515–5525. doi: 10.1158/1078-0432.CCR-05-0464. [DOI] [PubMed] [Google Scholar]
- 127.Fakhrai H, Mantil JC, Liu L, et al. Phase I clinical trial of a TGF-b antisense-modified tumor cell vaccine in patients with advanced glioma. Cancer Gene Ther. 2006;13:1052–1060. doi: 10.1038/sj.cgt.7700975. [DOI] [PubMed] [Google Scholar]
- 128.Butowski N, Chang SM, Junck L, et al. A phase II clinical trial of poly-ICLC with radiation for adult patients with newly diagnosed supratentorial glioblastoma: a North American Brain Tumor Consortium (NABTC01–05) J Neurooncol. 2009;91:175–182. doi: 10.1007/s11060-008-9693-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Sloan AE, Dansey R, Zamorano L, et al. Adoptive immunotherapy in patients with recurrent malignant glioma: preliminary results of using autologous whole-tumor vaccine plus granulocyte-macrophage colony-stimulating factor and adoptive transfer of anti-CD3-activated lymphocytes. Neurosurg Focus. 2000;9:e9. doi: 10.3171/foc.2000.9.6.10. [DOI] [PubMed] [Google Scholar]
- 130.Disis ML, Schiffman K, Gooley TA, McNeel DG, Rinn K, Knutson KL. Delayed-type hypersensitivity response is a predictor of peripheral blood T-cell immunity after HER-2/neu peptide immunization. Clin Cancer Res. 2000;6:1347–1350. [PubMed] [Google Scholar]
- 131.McNeel DG, Schiffman K, Disis ML. Immunization with recombinant human granulocyte-macrophage colony-stimulating factor as a vaccine adjuvant elicits both a cellular and humoral response to recombinant human granulocyte-macrophage colony-stimulating factor. Blood. 1999;93:2653–2659. [PubMed] [Google Scholar]
- 132.Thurner B, Haendle I, Roder C, et al. Vaccination with mage-3A1 peptide-pulsed mature, monocyte-derived dendritic cells expands specific cytotoxic T cells and induces regression of some metastases in advanced stage IV melanoma. J Exp Med. 1999;190:1669–1678. doi: 10.1084/jem.190.11.1669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Altman J, Moss P, Goulder P, et al. Phenotypic analysis of antigen-specific T lymphocytes. Science. 1996;274:94–96. [PubMed] [Google Scholar]
- 134.Yee C, Savage PA, Lee PP, Davis MM, Greenberg PD. Isolation of high avidity melanoma-reactive CTL from heterogeneous populations using peptide-MHC tetramers. J Immunol. 1999;162:2227–2234. [PubMed] [Google Scholar]
- 135.Mackensen A, Veelken H, Lahn M, et al. Induction of tumor-specific cytotoxic T lymphocytes by immunization with autologous tumor cells and interleukin-2 gene transfected fibroblasts. J Mol Med. 1997;75:290–296. doi: 10.1007/s001090050114. [DOI] [PubMed] [Google Scholar]
- 136.Dunbar PR, Ogg GS, Chen J, Rust N, van der Bruggen P, Cerundolo V. Direct isolation, phenotyping and cloning of low-frequency antigen-specific cytotoxic T lymphocytes from peripheral blood. Curr Biol. 1998;8:413–416. doi: 10.1016/s0960-9822(98)70161-7. [DOI] [PubMed] [Google Scholar]
- 137.Pittet MJ, Valmori D, Dunbar PR, et al. High frequencies of naive Melan-A/MART-1-specific CD8(+) T cells in a large proportion of human histocompatibility leukocyte antigen (HLA)-A2 individuals. J Exp Med. 1999;190:705–715. doi: 10.1084/jem.190.5.705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Schmittel A, Keilholz U, Scheibenbogen C. Evaluation of the interferon-gamma ELISPOT-assay for quantification of peptide specific T lymphocytes from peripheral blood. J Immunol Methods. 1997;210:167–174. doi: 10.1016/s0022-1759(97)00184-1. [DOI] [PubMed] [Google Scholar]