The site of the Media Sphera Publishers contains materials intended solely for healthcare professionals.
By closing this message, you confirm that you are a certified medical professional or a student of a medical educational institution.

Chekhonin I.V.

V.P. Serbskiy National Medical Research Center of Psychiatry and Narcology, Moscow, Russia;
N.N. Burdenko National Medical Research Center of Neurosurgery, Moscow, Russia

Kobiakov G.L.

GBU NII neĭrokhirurgii im. N.N. Burdenko

Gurina O.I.

Laboratoriia neĭrokhimii otdela fundamental'noĭ i prikladnoĭ neĭrobiologii Gosudarstvennogo nauchnogo tsentra sotsial'noĭ i sudebnoĭ psikhiatrii im. V.P. Serbskogo

Dendritic cell vaccines in neurological oncology

Authors:

Chekhonin I.V., Kobiakov G.L., Gurina O.I.

More about the authors

Journal: Burdenko's Journal of Neurosurgery. 2020;84(1): 76‑85

Read: 8503 times


To cite this article:

Chekhonin IV, Kobiakov GL, Gurina OI. Dendritic cell vaccines in neurological oncology. Burdenko's Journal of Neurosurgery. 2020;84(1):76‑85. (In Russ., In Engl.)
https://doi.org/10.17116/neiro20208401176

Recommended articles:
Anti­depressants as addi­tional drugs for human brain gliomas. Burdenko's Journal of Neurosurgery. 2024;(6):97-102
Glymphatic system in health and disease: a narrative review. Burdenko's Journal of Neurosurgery. 2025;(4):112-118
Machine learning models for brain tumors differential diagnosis. Russian Journal of Preventive Medi­cine. 2025;(9):87-93

Abbreviations

BMCP — biomedical cell product

G-CSF — granulocyte colony-stimulating factor

GM-CSF — granulocyte-macrophage colony-stimulating factor

IL — interleukin

IFN — interferon

mRNA — matrix ribonucleic acid

TNF — tumor necrosis factor

CD — cluster of differentiation

HLA — human leukocyte antigen

MGMT — O6-methylguanine DNA methyltransferase

RANO — response assessment in neuro-oncology

iRANO — immunotherapy response assessment in neuro-oncology

DC — dendritic cells

Immunotherapy of neuro-oncological diseases is a rapidly developing direction of experimental and clinical oncology. Dendritic cells vaccine is one of the clearest examples of active immunotherapy. Dendritic cells are specialized human antigen-presenting cells. These cells form primary immune response, capture, process and present foreign antigens to effector immune cells. Therefore, antigen-specific immune response is triggered. This mechanism also exists in normal conditions. However, tumors usually inhibit maturation and function of dendritic cells [1]. The technology of dendritic cell vaccines is currently aimed at correction of defective dendritic cell system. This technology implies dendritic cells differentiation and maturation in vitro from autologous hematopoietic precursors for their subsequent injection into the body. DC maturation implies enhancement of their ability to activate lymphocytes and, accordingly, stimulate immune response [2]. Standard protocols for stimulation of DC maturation contain data on the need to add cytokines including granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-4 (IL-4) [3]. The key moment in maturation is sensitization of DCs with the corresponding (tumor) antigens during cultivation. This ensures further antigen-specific immune response. The earliest experimental works on dendritic cell therapy of neuro-oncological diseases were performed on mouse and rat models of gliomas [4, 5]. Clinical trials of DCs were also performed after a short period. Perspective results of some researches (for example, in onco-urology) is an important stimulus. Thus, analysis of dendritic cell vaccines is rightfully an important direction in the treatment of brain tumors [6].

RESULTS OF CLINICAL TRIALS DEVOTED TO DENDRITIC CELL VACCINES FOR NEURO-ONCOLOGICAL DISEASES

Dendritic cell therapy of gliomas in adults

Currently, the vast majority of clinical trials of therapeutic efficacy of DC as an adjuvant therapy are devoted to the treatment of malignant gliomas. Various antigens may be used for DC-based vaccines (antigens of tumor cell lysates and extracts, individual proteins and peptides, nucleic acids). However, the feasibility of using certain antigens is largely determined by their specific releasing by tumor tissue. Most often, intraoperative tumor tissue specimens are used as antigens for DC sensitization. It is possible to use several antigens from the “collection” of these tumors. Sensitization process itself usually involves an addition of antigens to DC culture and subsequent joint incubation. Other techniques for DC sensitizing are discussed below on specific examples.

The advantages of dendritic cell vaccines sensitized with tumor antigens are no need to identify the antigens and attractiveness of this method from the standpoint of personalized medicine. Scheffer et al. reported higher immunogenicity of tumor lysates inactivated by irradiation followed by apoptosis compared with tumor lysates inactivated by freezing and thawing followed by necrosis [7]. At the same time, there is an evidence of immunosuppressive effect of apoptotic cell antigens [8]. DCs are administered subcutaneously as a rule.

To date, some trials of phase I and II have been carried out. There are preliminary data of phase III studies. Some trials of phase I included control group and survival rates were evaluated. The vaccines based on DC sensitized by tumor lysate antigens were predominantly studied for recurrent glioblastoma in early clinical trials of phase I and II. Small sample size should be considered despite the tendency to better survival in the treatment group. Yu et al. (trial phase I) reported greater median survival after redo surgery for the first recurrence of glioblastoma (n=8, 33.25 months) compared with the control group (n=26, 7.5 months). There were 3 vaccine injections with an interval of 2 weeks in the study group. Immune response against some tumor-associated antigens has been demonstrated in some patients. Patients with anaplastic astrocytoma and tumors de novo were excluded from survival analysis. There were no data on other adjuvant therapies after surgery for recurrent tumor in the study group while radiotherapy was performed in the control group [9].

De Vleeschouwer et al. similarly analyzed larger sample of patients (n=56). Adults and children were enrolled. In contrast to previous trials, dendritic cells were additionally exposed to IL-1β, tumor necrosis factor-α (TNF-α) and prostaglandin E2 in addition to standard IL-4 and GM-CSF. These cytokines increase yield, maturation, motility and immunostimulating properties of dendritic cells [10]. According to this report, median recurrence-free survival after the second surgery was 3 months, median overall survival — 9.6 months. It is important to note that reduced interval between DC injections from 2 to 1 week within one month with subsequent monthly injections of tumor lysate and total resection of the neoplasm were associated with improved recurrence-free survival [11].

New data on safety and efficacy of DC-based vaccines in the treatment of recurrent glioma ensured analysis of this method in patients with high-grade gliomas de novo. Chang et al. (trial phase I/II) used dendritic cells sensitized by autologous tumor antigens after exposure of tumor cells with interferon-γ (IFN-γ), heat shock and radiation. The authors reported median survival of patients with newly diagnosed glioblastoma (n=8) about 12,7 months, in patients with recurrent glioblastoma (n=8) — 32.2 months (all patients with glioblastoma — near 17.3 months). In authors’ opinion, better survival in the second group was associated with earlier vaccination. Indeed, radiotherapy was immediately followed by vaccine administration in patients with newly diagnosed glioblastomas while radiotherapy was not performed in patients with recurrent tumors. Moreover, early recurrences after immunotherapy in patients with newly diagnosed glioblastoma could be associated with appearance of cellular clones with acquired mutations after radiotherapy. Therefore, these cells could have another antigenic pattern compared with cells used to sensitize dendritic cells [12].

One of the options for sensitizing DCs with a complete set of tumor antigens is creation of chimeras of tumor and dendritic cells (derivatives after fusion of both types of cells). Kikuchi et al. used polyethylene glycol to obtain chimeras of irradiated glioma cells and human DC [13]. The first administration of DC was carried out in 2 weeks after temozolomide chemotherapy onset in each of the study groups (phase I/II, glioblastoma de novo or recurrent tumor). Injections were repeated at least three times in each of the 28-day chemotherapy courses. Median of recurrence-free period was 10.3 and 18.3 months in patients with recurrent glioblastoma or glioblastoma de novo, respectively. Median of overall survival was 18 and 30.5 months, respectively. Immune response against WT-1 (Wilms tumor protein 1), gp100 and melanoma-associated MAGE-A3 antigens (chemoresistance antigens) was recorded in response to vaccination with chimeric cells [14].

In our country, research of immunotherapy with dendritic cell vaccines is presented by reports performed at the Polenov Russian Research Institute of Neurosurgery together with the Konstantinov St. Petersburg Institute of Nuclear Physics, Research Institute of Fundamental and Clinical Immunology of Siberian Branch of the RAS together with the Institute of Cytology and Genetics and the Research Institute of Traumatology and Orthopedics (Novosibirsk).

The authors from St. Petersburg used destruction of irradiated fragments of gliomas with subsequent pH-dependent cellular lysis to prepare sensitizing DC mixture. A total of 141 patients (2000—2009) were included in the study of dendritic cell vaccine and activated lymphocytes for the management of high-grade gliomas. Median overall survival of patients undergoing immunotherapy with standard treatment was 25 months, in the control group — 15 months. Median recurrence-free period was 12 and 8 months, respectively [15]. According to the later report, DC administration was followed by median survival of 32 (n=18) and 24 (n=11) months in patients with anaplastic astrocytoma and glioblastoma, respectively [16].

The researchers from Novosibirsk used GM-CSF and interferon (IFN)-α for DC maturation. Three was a slightly lower yield of maturated DCs in patients with high-grade gliomas. However, DCs sensitized with antigens of tumor tissue lysates and cultured in the presence of IFN-α and GM-CSF had similar functional activity compared with cells of healthy people. These DCs more significantly activated T-helpers [17, 18]. Administration of these cells was followed by higher survival of patients with anaplastic astrocytoma and glioblastoma [19].

Analysis of clinical and immunological correlations is a separate question in the study of dendritic cell therapy. Wheeler et al. analyzed immune response of patients considering interferon-γ mRNA (IFN-γ) release by mononuclear cells (study phase II). Overall survival of patients with glioblastoma significantly correlated with clear immune response. Moreover, median and 2-year survival were higher in the group of significant response (more than 1.5-fold augmentation of IFN-γ release) compared with patients with insignificant immune response [20]. Pellegatta et al. reported increased level of blood natural killers after dendritic cell therapy that was accompanied by improved median recurrence-free and overall survival [21].

Importantly, the peptides non-specifically isolated by acid elution were used for DC sensitization besides glioma lysates in early clinical trials devoted to DC therapy of glioblastoma. Yu et al. [22] and Liau et al. [23] reported significant increase of median survival in patients with glioblastoma compared with the control groups (15.2 and 8.5 months; 23.4 and 18.3 months, respectively). DC administration caused systemic (immune cells) and intracranial (tumor infiltration by lymphocytes) cytotoxic response.

Glioma is characterized by increased release of certain antigenic peptides which are absent in normal tissues. Careful selection of these peptides contributes to creation of dendritic cell vaccines with specified antitumor properties. An important step in the development of dendritic cell vaccines sensitized with peptides is isolation of the antigens. For example, some cytomegalovirus antigens are released by more than 90% of glioblastomas (up to 90% of glioblastomas are infected with this virus). However, release of these antigens is absent in normal brain tissue [24, 25]. The role of cytomegalovirus in development of glioblastoma is insufficiently confirmed [26, 27]. Nevertheless, potential role of this virus as a target for immunotherapy looks very attractive [28]. Nair et al. analyzed immunogenicity of cytomegalovirus antigens. The authors sensitized human DCs with mRNA of pp65 phosphoprotein and then studied their ability to activate T-lymphocytes. It was shown that DC-stimulated T-lymphocytes were able to lyse autologous cells of glioblastoma [29]. Subsequently, the same authors reported high survival in patients with glioblastoma de novo undergoing dendritic cell and temozolomide therapy after tumor resection and initial course of chemoradiotherapy. Median recurrence-free period was 25.3 months, median overall survival — 41.1 months. These values were higher compared with the control group despite the expansion of T-regulatory (“suppressor”) cells after therapy with temozolomide [25].

Phuphanich et al. used a mixture of antigen epitopes associated with gliomas and overexposed on the surface of tumor stem cells for DC sensitization (epidermal growth factor receptor HER2, glycoprotein gp100, MAGE-1, tyrosinase-related protein (TRP-2), subunit α2 of IL-13 receptor ( IL13Rα2), interferon-inducible protein — a component of inflammosomes (AIM-2)). Injection of this vaccine (ICT-107) reduced expression of glioma stem cell marker genes in tumor samples [30]. Application of sensitized DCs was accompanied by a tendency to increase of overall survival and significant prolongation of recurrence-free period [31]. In 2016, Phuphanich et al. reported 6 patients with more than 6-year survival and 3 patients with more than 8-year survival in the treatment group [32].

Unification of vaccination conditions and greater sample size in clinical trial phase III were necessary to determine the effect of vaccines on survival rates. Currently, these tests are conducted regarding the technology of manufacturing an autologous dendritic cell vaccine DCVax. Antigens of autologous glial tumor lysates are used to sensitize DCs in this vaccine [33]. Liau et al. reported intermediate results of DCVax vaccine administration. Overall group enrolled 331 patients. Side effects grade 3—4 (National Cancer Institute Common Toxicity Criteria) at least presumably associated with therapy were observed in 7 patients (cerebral edema, seizures, severe nausea, lymphadenitis). The authors reported satisfactory tolerability of the vaccine and increased survival compared to standard therapeutic approaches for glioblastoma de novo. Median overall survival was 23.1 months, in patients with methylated MGMT gene promoter (O6-methylguanine DNA methyltransferase gene) — 34.7 months after surgery, 3-year survival — 46.4%. However, there were small subgroups of patients with median survival of 46.5 and 88.2 months, respectively [34]. Moreover, ICT-107 vaccine (NCT02546102) is currently under investigation (study phase III).

Dendritic cell therapy of intracerebral tumors in children

Clinical trials of immunotherapy in children are less developed despite successful management of brain tumors in adults. In part, this may be due to sampling of insufficient volume of tumor tissue, for example, in patients with pontine gliomas. However, Boczkowski et al. reported that about 2000 tumor cells are sufficient for amplification of the required amount of RNA in order to create a potentially immunogenic vaccine [35]. Another common problem is impaired function of mononuclear cells after cytotoxic therapy (radio- or chemotherapy) and actually on the background of tumor process that reduces yield of dendritic cells [36]. There is a technique of administering granulocyte growth factor (G-CSF) prior to obtaining mononuclear fraction in order to increase yield of dendritic cells [37].

There is evidence of obtaining blood DCs from the patients with medulloblastoma accounting up to 20% of brain tumors in children [38].

Up to 1/3 of tumors are characterized by recurrent course despite various modern methods of therapy [39]. Moreover, 3-year survival of patients with recurrent medulloblastoma is less than 20% [40]. Nair et al. reported fundamental possibility to increase yield and quality of mononuclear precursors of dendritic cells in children after chemotherapy and subsequent induction with G-CSF. Monocytic CD14+ fraction was used for differentiation of dendritic cells. DC electroporation was performed using plasmids with RNA of some proteins (matrix protein of Flu M1 virus, cytomegalovirus pp65 protein or survivin protein). Plasmids were injected through the pores in the cellular membrane created under electric field. DC maturation was achieved in the presence of TNF-α, IL-1β, IL-6, prostaglandin E2. Adequate mobilization of monocytes for differentiation into DK was observed in 4 out of 5 patients while proper amount of mature phenotype of dendritic cells was obtained in 3 out of these 4 patients. However, functional activity of DC was absent in one patient due to insufficient mobilization of T-lymphocytes. Thus, the authors reported the possibility to obtain functionally active DCs from patients with refractory medulloblastoma after chemotherapy [41].

There are attempts to use dendritic cell vaccines in patients with diffuse pontine glioma. This malignancy (grade IV) is referred to diffuse midline gliomas in accordance with 2016 WHO classification [42]. Immunotherapy of this neoplasm is justified by specific mutation of H3 histone associated with replacement of lysine with methionine at the position 27 (K27M). This mutation determines potential immunogenicity of H3 histone [43]. A peptide vaccine triggering immune response against above-mentioned epitope (NCT02960230) is currently being investigated. However, there are no data on the use of this antigen for DC sensitization.

Benitez-Ribas et al. obtained autologous DCs from mononuclear cells of children with diffuse stem glioma. Dendritic cells were sensitized with antigens of cellular culture of this glioma. Specific immunogenicity and safety of dendritic cell vaccine were reported in 8 out of 9 patients (with different H3 K27M status) [44].

Research of the effectiveness of DC-based vaccines sensitized with tumor RNA and adaptive lymphocyte transfer for recurrent medulloblastoma and primitive neuroectodermal tumor is currently ongoing (NCT01326104). Ardon et al. analyzed children with high-grade glioma, medulloblastoma, primitive neuroectodermal tumor, ependymoma and atypical teratoid-rhabdoid tumor. The authors found more significant immune response in patients with glioma and atypical teratoid-rhabdoid tumor compared with medulloblastoma, primitive neuroepithelial tumor and ependymoma [45].

It should be noted that gliomas in children differ by another pattern of tumor antigen expression compared with adults. At the same time, cytomegalovirus gene expression is also increased in childhood gliomas that makes these antigens a perspective target for therapy in pediatric neuro-oncology [46]. Nevertheless, the question active immunotherapy for gliomas in children remains unclear [47].

Dendritic cell therapy of intracerebral metastases

Effective treatment of metastatic tumors is a very difficult task. Dendritic cell therapy may be applied in these patients. Brain metastases was an exclusion criterion in a large number of trials devoted to immunotherapy [48].

Clinical data on the efficacy of dendritic cell vaccines in the treatment of intracerebral metastases are presented by small studies and case reports. There is evidence of a positive interaction between radiotherapy and immunotherapy in patients with brain metastases of melanoma. Karbach et al. reported a patient with similar metastases and 10-year remission after radiotherapy and DC vaccination. Autologous dendritic cells were sensitized with antigens of metastatic tumor tissue lysate obtained from lymph nodes. There was also immune response in the form of expansion of CD8+ T cells specific for MAGE-A1 antigen of melanoma [49].

Dillman et al. demonstrated safety of autologous combination of GM-CSF and dendritic cell vaccine. Dendritic cells were sensitized with antigens of previously cultured in the presence of IFN-γ, irradiated and frozen cells of melanoma. Brain metastases were diagnosed in 3 patients of the experimental group. Individual calculations of survival were not performed in this group. Median overall survival was 13.8 months [50].

Mayordomo et al. reported clinical improvement for 8 months in a patient with skin, liver, lung and bone metastases of melanoma after administration of dendritic cells. However, brain metastasis was detected later [51]. Laurell et al. injected allogeneic DC in the primary tumor and reported the effect of dendritic cell therapy on regression of intracerebral metastases. Patients with metastatic renal cell carcinoma were predominantly analyzed. Allogeneic DCs were used due to possible development of bystander effect. It is enhanced immune response due to functioning of injected DCs and immune induction associated with DC non-sensitized with certain antigen. Patients with intracerebral metastases were excluded. However, there was a patient with intracerebral metastases occurred after therapy onset. Their regression was further noted. It should be additionally noted that this patient received tyrosine kinase inhibitor (sunitinib) [52].

Okwan-Duodu et al. reported high efficacy of radiotherapy combined with IL-2 immunotherapy for intracerebral metastases of melanoma. The possible mechanism of this effect is explained by potentiation of dendritic-cell unit after post-radiation release of melanoma-associated antigens.

Thus, combination of the above-mentioned treatment with additional administration of dendritic cell vaccines seems to be very interesting and requiring further analysis [53].

Actual issues of dendritic cell therapy of gliomas

Currently, there are several unresolved issues regarding the future of dendritic cell vaccines in clinical practice. Other methods of active immunotherapy are being studied in parallel with creation of these vaccines (for example, vaccination of patients with immunogenic peptides without DCs).

In 2017, Weller et al. published a review devoted to fundamental and applied aspects of immunotherapy for glioblastoma [54]. The authors supposed that successful antitumor vaccination may be achieved by combinations of these strategies with targeted therapy, for example, monoclonal antibodies (modulators of immune response control points) and immunosuppressive metabolic pathway inhibitors. Weller et al. emphasize the concept of relationship of response to therapy with genotype. Indeed, this approach reflects personification as a trend in translational medicine. For example, potential association of effective therapy with HLA-A2 allele was shown in the above-mentioned study of ICT-107 vaccine [30]. Only HLA-A2-positive patients were enrolled in the NCT02546102 study. Some trials of dendritic cell vaccines and MGMT status found higher survival in the group with methylated MGMT promoter [34, 55]. In our opinion, correlation of MGMT promoter methylation with successful dendritic cell therapy requires confirmation, since this prognostically favorable factor and immunotherapy can act independently of each other. However, Liau et al. supposed correlation between the effect of dendritic cell vaccine and temozolomide therapy [34].

A very important aspect of clinical research is integration of dendritic cell therapy into the overall treatment plan. Features of interaction of dendritic cell vaccines with systemic administration of corticosteroids are unclear while corticosteroids are one of the main components of symptomatic therapy for intracerebral tumor. As a rule, high-dose corticosteroid therapy was an exclusion criterion considering significant immunosuppressive effect. Any prescription of corticosteroids was an exclusion criterion in some studies [11, 56], while limitation of doses was possible in other ones [12]. Some earlier researches in vitro did not show a significant effect of dexamethasone on DC maturation per se and their phagocytic activity [57]. Further studies demonstrated immunosuppressive effect of this drug due to a special pool of monocytes [58].

Adaptation of therapeutic response criteria in glioma patients with particular emphasis on immunotherapy is also significant. New RANO criteria were published in 2015 (Response Assessment in Neuro-Oncology, Okada et al.) which were called iRANO (immunotherapy RANO) [59]. Some features of tumor response against this type of therapy were taken into account during development of new criteria. For example, pseudoprogression following immunotherapy can develop later compared with standard therapy (6 vs. 3 months). Moreover, immunotherapy may be followed by response after appearance of visual signs of tumor progression. Thus, it is not established whether continuation of immunotherapy in patients with confirmed tumor progression is possible. In some cases, immunotherapeutic agents can initiate inflammatory response. These inflammatory infiltrates can mimic visual signs of tumor progression including typical contrast enhancement. Obviously, various immunotherapeutic methods probably imply different response patterns that can require additional refinement of the criteria.

An important current problem is organization of clinical trials of dendritic cell vaccines in Russia. The authors of the aforementioned national trials talk not so much a clinical study but intermediate nature of the work between research and application of dendritic cell vaccines. For example, it is unclear what patients were analyzed in these reports (tumor de novo or recurrence). There is also no clear analysis of toxicity and safety of the drugs (except for general formulations about the absence of “serious complications”). Federal Law No. 180-FZ “On Biomedical Cellular Products” dated on June 23, 2016 has entered into force since January 1, 2017. Chapter 5 of the Article 35 of this law (entered into force on January 1, 2018) regulates manufacturing of biomedical cellular preparations exclusively in accordance with Good Clinical Practice guidelines (approved by the order of the Ministry of Health of the Russian Federation No. 669n on September 22, 2017). In our opinion, correspondence of dendritic cell vaccines to the concept of “biomedical cellular preparations” is relevant issue. According to definition, this product should be a “standardized population of cells with reproducible cellular composition” obtained by cultivation. However, maturation of dendritic cells includes their differentiation from the precursors. Therefore, lost ability of cellular reproduction is implied. At the same time, modern legal practice considers dendritic-cell vaccines exactly as biomedical cellular preparations that requires additional clarifications in order to avoid legal conflicts.

Conclusion

We considered several approaches to clinical application of dendritic cell vaccines. The results of some clinical trials are presented. It is known about the planning of new studies. Safety of vaccines based on autologous dendritic cells and small incidence of adverse side effects in a large percentage of cases have been reported in various researches since 2000. Analysis of various dendritic cell vaccination strategies in the treatment of gliomas resulted a tendency to increased survival rates. It is difficult to determine the absolute advantages of a particular vaccination strategy considering scientific data in trial phases I and II, as well as preliminary results of studies phase III because different methods and design of various clinical studies are characterized by various advantages and disadvantages. At the same time, there is a tendency towards some positive effects of dendritic cell therapy. This tendency includes additional immune response induced by vaccines and mild improvement of survival. Comprehensive assessment and publication of the final results of the studies phase III is planned in the near future. These data will be valuable to determine the role of dendritic cell vaccines in neuro-oncology.

Authors’ participation

Concept and design of the study — I.CH., G.K., O.G.

Collection and analysis of data — I.CH., G.K., O.G.

Writing the text — I.CH., G.K., O.G.

Editing — G.K., O.G.

The authors declare no conflicts of interest.

References:

  1. Wang J, Liu P, Xin S, Wang Z, Li J. Nrf2 suppresses the function of dendritic cells to facilitate the immune escape of glioma cells. Experimental Cell Research. 2017;360(2):66-73. https://doi.org/10.1016/j.yexcr.2017.07.031
  2. Boyette LB, Macedo C, Hadi K, Elinoff BD, Walters JT, Ramaswami B, Chalasani G, Taboas JM, Lakkis FG, Metes DM. Phenotype, function, and differentiation potential of human monocyte subsets. PLoS One. 2017;12(4):e0176460. https://doi.org/10.1371/journal.pone.0176460
  3. Hochrein H, O’Keeffe M, Luft T, Vandenabeele S, Grumont RJ, Maraskovsky E, Shortman K. Interleukin (IL)-4 is a major regulatory cytokine governing bioactive IL-12 production by mouse and human dendritic cells. Journal of Experimental Medicine. 2000;192(6):823-833.
  4. Liau LM, Black KL, Prins RM, Sykes SN, DiPatre PL, Cloughesy TF, Becker DP, Bronstein JM. Treatment of intracranial gliomas with bone marrow-derived dendritic cells pulsed with tumor antigens. Journal of Neurosurgery. 1999;90(6):1115-1124. https://doi.org/10.3171/jns.1999.90.6.1115
  5. Siesjo P, Visse E, Sjogren HO. Cure of established, intracerebral rat gliomas induced by therapeutic immunizations with tumor cells and purified APC or adjuvant IFN-gamma treatment. Journal of Immunotherapy with Emphasis on Tumor Immunology. 1996;19(5):334-345.
  6. Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, Redfern CH, Ferrari AC, Dreicer R, Sims RB, Xu Y, Frohlich MW, Schellhammer PF. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. New England Journal of Medicine. 2010;363(5):411-422. https://doi.org/10.1056/NEJMoa1001294
  7. Scheffer SR, Nave H, Korangy F, Schlote K, Pabst R, Jaffee EM, Manns MP, Greten TF. Apoptotic, but not necrotic, tumor cell vaccines induce a potent immune response in vivo. International Journal of Cancer. 2003;103(2):205-211. https://doi.org/10.1002/ijc.10777
  8. Linke B, Abeler-Dorner L, Jahndel V, Kurz A, Mahr A, Pfrang S, Linke L, Krammer PH, Weyd H. The tolerogenic function of annexins on apoptotic cells is mediated by the annexin core domain. Journal of Immunology. 2015;194(11):5233-5242. https://doi.org/10.4049/jimmunol.1401299
  9. Yu JS, Liu G, Ying H, Yong WH, Black KL, Wheeler CJ. Vaccination with tumor lysate-pulsed dendritic cells elicits antigen-specific, cytotoxic T-cells in patients with malignant glioma. Cancer Research. 2004;64(14):4973-4979. https://doi.org/10.1158/0008-5472.can-03-3505
  10. Jonuleit H, Kuhn U, Muller G, Steinbrink K, Paragnik L, Schmitt E, Knop J, Enk AH. Pro-inflammatory cytokines and prostaglandins induce maturation of potent immunostimulatory dendritic cells under fetal calf serum-free conditions. European Journal of Immunology. 1997;27(12):3135-3142. https://doi.org/10.1002/eji.1830271209
  11. De Vleeschouwer S, Fieuws S, Rutkowski S, Van Calenbergh F, Van Loon J, Goffin J, Sciot R, Wilms G, Demaerel P, Warmuth-Metz M, Soerensen N, Wolff JE, Wagner S, Kaempgen E, Van Gool SW. Postoperative adjuvant dendritic cell-based immunotherapy in patients with relapsed glioblastoma multiforme. Clinical Cancer Research. 2008;14(10):3098-3104. https://doi.org/10.1158/1078-0432.ccr-07-4875
  12. Chang CN, Huang YC, Yang DM, Kikuta K, Wei KJ, Kubota T, Yang WK. A phase I/II clinical trial investigating the adverse and therapeutic effects of a postoperative autologous dendritic cell tumor vaccine in patients with malignant glioma. Journal of Clinical Neuroscience. 2011;18(8):1048-1054. https://doi.org/10.1016/j.jocn.2010.11.034
  13. Kikuchi T, Akasaki Y, Irie M, Homma S, Abe T, Ohno T. Results of a phase I clinical trial of vaccination of glioma patients with fusions of dendritic and glioma cells. Cancer Immunology and Immunotherapy. 2001;50(7):337-344. https://doi.org/10.1007/s002620100205
  14. Akasaki Y, Kikuchi T, Homma S, Koido S, Ohkusa T, Tasaki T, Hayashi K, Komita H, Watanabe N, Suzuki Y, Yamamoto Y, Mori R, Arai T, Tanaka T, Joki T, Yanagisawa T, Murayama Y. Phase I/II trial of combination of temozolomide chemotherapy and immunotherapy with fusions of dendritic and glioma cells in patients with glioblastoma. Cancer Immunology and Immunotherapy. 2016;65(12):1499-1509. https://doi.org/10.1007/s00262-016-1905-7
  15. Olyushin VE, Filatov MV, Ulitin AYu, Rostovtsev DM, Fadeyeva TN, Maslova LN, Papayan GV. New technologies in therapy of patients with malignant gliomas of cerebral hemispheres. Prakticheskaya onkologiya. 2013;14(3):175-179. (In Russ.)
  16. Olyushin VE, Rostovtsev DM, Papayan GV, Filatov MV, Fadeyeva TN. Photodynamic therapy and specific antitumor immune therapy in the structure of integrated treatment of patients with malignant astrocytic supratentorial tumors. Long-term treatment results. Medicinskij vestnik Yuga Rossii. 2014;4:83-89. (In Russ.)
  17. Leplina OYu, Tihonova MA, Kozlov YuP, Stupak VV, Nikonov SD, Ostanin AA, Chernykh ER. Semi-mature dendritic cells as a potential basis for the induction of anti-tumor response in patients with malignant gliomas. Medicinskaya immunologiya. 2005;7(4):365-374. (In Russ.) https://doi.org/10.15789/1563-0625-2005-4-365-374
  18. Leplina OYu, Tikhonova MA, Tyrinova TV, Alyamkina EA, Bogachev SS, Ostanin AA, Chernykh ER. Functional activity of IFNα- and IL-4-induced human dendritic cells: A comparative study. Medicinskaya immunologiya. 2014;16(1):43-52. (In Russ.) https://doi.org/10.15789/1563-0625-2014-1-43-52
  19. Leplina OYu, Stupak VV, Kozlov YuP, Pendyurin IV, Nikonov SD, Tikhonova MA, Sycheva NV, Ostanin AA, Chernykh ER. Use of interferon-α-induced dendritic cells in the therapy of patients with malignant brain gliomas. Kletochnye tekhnologii v biologii i medicine. 2007;2:92-98. (In Russ.) https://doi.org/10.1007/s10517-007-0172-1
  20. Wheeler CJ, Black KL, Liu G, Mazer M, Zhang XX, Pepkowitz S, Goldfinger D, Ng H, Irvin D, Yu JS. Vaccination elicits correlated immune and clinical responses in glioblastoma multiforme patients. Cancer Research. 2008;68(14):5955-5964. https://doi.org/10.1158/0008-5472.can-07-5973
  21. Pellegatta S, Eoli M, Frigerio S, Antozzi C, Bruzzone MG, Cantini G, Nava S, Anghileri E, Cuppini L, Cuccarini V, Ciusani E, Dossena M, Pollo B, Mantegazza R, Parati EA, Finocchiaro G. The natural killer cell response and tumor debulking are associated with prolonged survival in recurrent glioblastoma patients receiving dendritic cells loaded with autologous tumor lysates. Oncoimmunology. 2013;2(3):e23401. https://doi.org/10.4161/onci.23401
  22. Yu JS, Wheeler CJ, Zeltzer PM, Ying H, Finger DN, Lee PK, Yong WH, Incardona F, Thompson RC, Riedinger MS, Zhang W, Prins RM, Black KL. Vaccination of malignant glioma patients with peptide-pulsed dendritic cells elicits systemic cytotoxicity and intracranial T-cell infiltration. Cancer Research. 2001;61(3):842-847.
  23. Liau LM, Prins RM, Kiertscher SM, Odesa SK, Kremen TJ, Giovannone AJ, Lin JW, Chute DJ, Mischel PS, Cloughesy TF, Roth MD. Dendritic cell vaccination in glioblastoma patients induces systemic and intracranial T-cell responses modulated by the local central nervous system tumor microenvironment. Clinical Cancer Research. 2005;11(15):5515-5525. https://doi.org/10.1158/1078-0432.ccr-05-0464
  24. Cobbs CS, Harkins L, Samanta M, Gillespie GY, Bharara S, King PH, Nabors LB, Cobbs CG, Britt WJ. Human cytomegalovirus infection and expression in human malignant glioma. Cancer Research. 2002;62(12):3347-3350.
  25. Batich KA, Reap EA, Archer GE, Sanchez-Perez L, Nair SK, Schmittling RJ, Norberg P, Xie W, Herndon JE, 2nd, Healy P, McLendon RE, Friedman AH, Friedman HS, Bigner D, Vlahovic G, Mitchell DA, Sampson JH. Long-term Survival in Glioblastoma with Cytomegalovirus pp65-Targeted Vaccination. Clinical Cancer Research. 2017;23(8):1898-1909. https://doi.org/10.1158/1078-0432.ccr-16-2057
  26. Dziurzynski K, Chang SM, Heimberger AB, Kalejta RF, McGregor Dallas SR, Smit M, Soroceanu L, Cobbs CS. Consensus on the role of human cytomegalovirus in glioblastoma. Neuro-Oncology. 2012;14(3):246-255. https://doi.org/10.1093/neuonc/nor227
  27. Dey M, Ahmed AU, Lesniak MS. Cytomegalovirus and glioma: putting the cart before the horse. Journal of Neurology, Neurosurgery, and Psychiatry. 2015;86(2):191-199. https://doi.org/10.1136/jnnp-2014-307727
  28. Sampson JH, Mitchell DA. Is cytomegalovirus a therapeutic target in glioblastoma? Clinical Cancer Research. 2011;17(14):4619-4621. https://doi.org/10.1158/1078-0432.ccr-11-0992
  29. Nair SK, De Leon G, Boczkowski D, Schmittling R, Xie W, Staats J, Liu R, Johnson LA, Weinhold K, Archer GE, Sampson JH, Mitchell DA. Recognition and killing of autologous, primary glioblastoma tumor cells by human cytomegalovirus pp65-specific cytotoxic T cells. Clinical Cancer Research. 2014;20(10):2684-2694. https://doi.org/10.1158/1078-0432.ccr-13-3268
  30. Phuphanich S, Wheeler CJ, Rudnick JD, Mazer M, Wang H, Nuno MA, Richardson JE, Fan X, Ji J, Chu RM, Bender JG, Hawkins ES, Patil CG, Black KL, Yu JS. Phase I trial of a multi-epitope-pulsed dendritic cell vaccine for patients with newly diagnosed glioblastoma. Cancer Immunology and Immunotherapy. 2013;62(1):125-135. https://doi.org/10.1007/s00262-012-1319-0
  31. Wen PY. A randomized, double blind, placebo-controlled phase 2 trial of dendritic cell (DC) vaccination with ICT-107 in newly diagnosed glioblastoma (GBM) patients. Neuro-Oncology. 2014;16(suppl 5):22. https://doi.org/10.1093/neuonc/nou237.59
  32. Phuphanich S, Wheeler C, Rudnick J, Hu J. Long term remission/survival over 8 years in patients with newly diagnosed glioblastoma (GBM) treated with ICT-107 dendritic cell-based immunotherapy (phase I). Neuro-Oncology. 2016;18(suppl 4):iv5. https://doi.org/10.1093/neuonc/now188.015
  33. Polyzoidis S, Ashkan K. DCVax(R)-L — developed by Northwest Biotherapeutics. Human Vaccines and Immunotherapeutics. 2014;10(11):3139-3145. https://doi.org/10.4161/hv.29276
  34. Liau LM, Ashkan K, Tran DD, Campian JL, Trusheim JE, Cobbs CS, Heth JA, Salacz M, Taylor S, D’Andre SD, Iwamoto FM, Dropcho EJ, Moshel YA, Walter KA, Pillainayagam CP, Aiken R, Chaudhary R, Goldlust SA, Bota DA, Duic P, Grewal J, Elinzano H, Toms SA, Lillehei KO, Mikkelsen T, Walbert T, Abram SR, Brenner AJ, Brem S, Ewend MG, Khagi S, Portnow J, Kim LJ, Loudon WG, Thompson RC, Avigan DE, Fink KL, Geoffroy FJ, Lindhorst S, Lutzky J, Sloan AE, Schackert G, Krex D, Meisel HJ, Wu J, Davis RP, Duma C, Etame AB, Mathieu D, Kesari S, Piccioni D, Westphal M, Baskin DS, New PZ, Lacroix M, May SA, Pluard TJ, Tse V, Green RM, Villano JL, Pearlman M, Petrecca K, Schulder M, Taylor LP, Maida AE, Prins RM, Cloughesy TF, Mulholland P, Bosch ML. First results on survival from a large Phase 3 clinical trial of an autologous dendritic cell vaccine in newly diagnosed glioblastoma. Journal of Translational Medicine. 2018;16(1):142. https://doi.org/10.1186/s12967-018-1507-6
  35. Boczkowski D, Nair SK, Nam JH, Lyerly HK, Gilboa E. Induction of tumor immunity and cytotoxic T lymphocyte responses using dendritic cells transfected with messenger RNA amplified from tumor cells. Cancer Research. 2000;60(4):1028-1034.
  36. Vakkila J, Vettenranta K, Sariola H, Saarinen-Pihkala UM. Poor yield of dendritic cell precursors from untreated pediatric cancer. Journal of Hematotherapy and Stem Cell Research. 2001;10(6):787-793. https://doi.org/10.1089/152581601317210881
  37. Paczesny S, Li YP, Li N, Latger-Cannard V, Marchal L, Ou-Yang JP, Bordigoni P, Stoltz JF, Eljaafari A. Efficient generation of CD34+ progenitor-derived dendritic cells from G-CSF-mobilized peripheral mononuclear cells does not require hematopoietic stem cell enrichment. Journal of Leukocyte Biology. 2007;81(4):957-967. https://doi.org/10.1189/jlb.0406296
  38. Khanna V, Achey RL, Ostrom QT, Block-Beach H, Kruchko C, Barnholtz-Sloan JS, de Blank PM. Incidence and survival trends for medulloblastomas in the United States from 2001 to 2013. Journal of Neuro-Oncology. 2017;135(3):433-441. https://doi.org/10.1007/s11060-017-2594-6
  39. Johnston DL, Keene D, Strother D, Taneva M, Lafay-Cousin L, Fryer C, Scheinemann K, Carret AS, Fleming A, Afzal S, Wilson B, Bowes L, Zelcer S, Mpofu C, Silva M, Larouche V, Brossard J, Bouffet E. Survival Following Tumor Recurrence in Children With Medulloblastoma. Journal of Pediatric Hematology/Oncology. 2018;40(3):159-163. https://doi.org/10.1097/mph.0000000000001095
  40. Koschmann C, Bloom K, Upadhyaya S, Geyer JR, Leary SE. Survival After Relapse of Medulloblastoma. Journal of Pediatric Hematology/Oncology. 2016;38(4):269-273. https://doi.org/10.1097/mph.0000000000000547
  41. Nair SK, Driscoll T, Boczkowski D, Schmittling R, Reynolds R, Johnson LA, Grant G, Fuchs H, Bigner DD, Sampson JH, Gururangan S, Mitchell DA. Ex vivo generation of dendritic cells from cryopreserved, post-induction chemotherapy, mobilized leukapheresis from pediatric patients with medulloblastoma. Journal of Neuro-Oncology. 2015;125(1):65-74. https://doi.org/10.1007/s11060-015-1890-2
  42. Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues P, Ellison DW. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathologica. 2016;131(6):803-820. https://doi.org/10.1007/s00401-016-1545-1
  43. Ochs K, Ott M, Bunse T, Sahm F, Bunse L, Deumelandt K, Sonner JK, Keil M, von Deimling A, Wick W, Platten M. K27M-mutant histone-3 as a novel target for glioma immunotherapy. Oncoimmunology. 2017;6(7):e1328340. https://doi.org/10.1080/2162402x.2017.1328340
  44. Benitez-Ribas D, Cabezon R, Florez-Grau G, Molero MC, Puerta P, Guillen A, Paco S, Carcaboso AM, Santa-Maria Lopez V, Cruz O, de Torres C, Salvador N, Juan M, Mora J, La Madrid AM. Immune Response Generated with the Administration of Autologous Dendritic Cells Pulsed with an Allogenic Tumoral Cell-Lines Lysate in Patients with Newly Diagnosed Diffuse Intrinsic Pontine Glioma. Frontiers in Oncolology. 2018;8:127. https://doi.org/10.3389/fonc.2018.00127
  45. Ardon H, De Vleeschouwer S, Van Calenbergh F, Claes L, Kramm CM, Rutkowski S, Wolff JE, Van Gool SW. Adjuvant dendritic cell-based tumour vaccination for children with malignant brain tumours. Pediatric Blood and Cancer. 2010;54(4):519-525. https://doi.org/10.1002/pbc.22319
  46. Wakefield A, Pignata A, Ghazi A, Ashoori A, Hegde M, Landi D, Gray T, Scheurer ME, Chintagumpala M, Adesina A, Gottschalk S, Hicks J, Powell SZ, Ahmed N. Is CMV a target in pediatric glioblastoma? Expression of CMV proteins, pp65 and IE1-72 and CMV nucleic acids in a cohort of pediatric glioblastoma patients. Journal of Neuro-Oncology. 2015;125(2):307-315. https://doi.org/10.1007/s11060-015-1905-z
  47. Rizzo D, Ruggiero A, Martini M, Rizzo V, Maurizi P, Riccardi R. Molecular Biology in Pediatric High-Grade Glioma: Impact on Prognosis and Treatment. BioMed Research International. 2015;215135. https://doi.org/10.1155/2015/215135
  48. Amin A, Dudek AZ, Logan TF, Lance RS, Holzbeierlein JM, Knox JJ, Master VA, Pal SK, Miller WH, Jr, Karsh LI, Tcherepanova IY, DeBenedette MA, Williams WL, Plessinger DC, Nicolette CA, Figlin RA. Survival with AGS-003, an autologous dendritic cell-based immunotherapy, in combination with sunitinib in unfavorable risk patients with advanced renal cell carcinoma (RCC): Phase 2 study results. Journal for ImmunoTherapy of Cancer. 2015;3:14. https://doi.org/10.1186/s40425-015-0055-3
  49. Karbach J, Gnjatic S, Biskamp M, Atmaca A, Weidmann E, Brandt K, Wahle C, Bernhard H, Knuth A, Jager E. Long-term complete remission following radiosurgery and immunotherapy in a melanoma patient with brain metastasis: immunologic correlates. Cancer Immunology Research. 2014;2(5):404-409. https://doi.org/10.1158/2326-6066.cir-13-0200
  50. Dillman R, Selvan S, Schiltz P, Peterson C, Allen K, Depriest C, McClay E, Barth N, Sheehy P, de Leon C, Beutel L. Phase I/II trial of melanoma patient-specific vaccine of proliferating autologous tumor cells, dendritic cells, and GM-CSF: planned interim analysis. Cancer Biotherapy and Radiopharmaceuticals. 2004;19(5):658-665. https://doi.org/10.1089/cbr.2004.19.658
  51. Mayordomo JI, Andres R, Isla MD, Murillo L, Cajal R, Yubero A, Blasco C, Lasierra P, Palomera L, Fuertes MA, Guemes A, Sousa R, Garcia-Prats M, Escudero P, Saenz A, Godino J, Marco I, Saez B, Visus C, Asin L, Valdivia G, Larrad L, Tres A. Results of a pilot trial of immunotherapy with dendritic cells pulsed with autologous tumor lysates in patients with advanced cancer. Tumori. 2007;93(1):26-30. https://doi.org/10.1177/030089160709300106
  52. Laurell A, Lonnemark M, Brekkan E, Magnusson A, Tolf A, Wallgren AC, Andersson B, Adamson L, Kiessling R, Karlsson-Parra A. Intratumorally injected pro-inflammatory allogeneic dendritic cells as immune enhancers: a first-in-human study in unfavourable risk patients with metastatic renal cell carcinoma. Journal for ImmunoTherapy of Cancer. 2017;5:52. https://doi.org/10.1186/s40425-017-0255-0
  53. Okwan-Duodu D, Pollack BP, Lawson D, Khan MK. Role of radiation therapy as immune activator in the era of modern immunotherapy for metastatic malignant melanoma. American Journal of Clinical Oncology. 2015;38(1):119-125. https://doi.org/10.1097/COC.0b013e3182940dc3
  54. Weller M, Roth P, Preusser M, Wick W, Reardon DA, Platten M, Sampson JH. Vaccine-based immunotherapeutic approaches to gliomas and beyond. Nature Reviews Neurology. 2017;13(6):363-374. https://doi.org/10.1038/nrneurol.2017.64
  55. Inoges S, Tejada S, de Cerio AL, Gallego Perez-Larraya J, Espinos J, Idoate MA, Dominguez PD, de Eulate RG, Aristu J, Bendandi M, Pastor F, Alonso M, Andreu E, Cardoso FP, Valle RD. A phase II trial of autologous dendritic cell vaccination and radiochemotherapy following fluorescence-guided surgery in newly diagnosed glioblastoma patients. Journal of Translational Medicine. 2017;15(1):104. https://doi.org/10.1186/s12967-017-1202-z
  56. Vik-Mo EO, Nyakas M, Mikkelsen BV, Moe MC, Due-Tonnesen P, Suso EM, Saeboe-Larssen S, Sandberg C, Brinchmann JE, Helseth E, Rasmussen AM, Lote K, Aamdal S, Gaudernack G, Kvalheim G, Langmoen IA. Therapeutic vaccination against autologous cancer stem cells with mRNA-transfected dendritic cells in patients with glioblastoma. Cancer Immunology and Immunotherapy. 2013;62(9):1499-1509. https://doi.org/10.1007/s00262-013-1453-3
  57. Takagi Y, Kikuchi T, Niimura M, Ohno T. Effects of glucocorticoids on antitumor effects of immunizations with fusions of dendritic and tumor cells. Anticancer Research. 2003;23(3B):2553-2558.
  58. Gustafson MP, Lin Y, New KC, Bulur PA, O’Neill BP, Gastineau DA, Dietz AB. Systemic immune suppression in glioblastoma: the interplay between CD14+HLA-DRlo/neg monocytes, tumor factors, and dexamethasone. Neuro-Oncology. 2010;12(7):631-644. https://doi.org/10.1093/neuonc/noq001
  59. Okada H, Weller M, Huang R, Finocchiaro G, Gilbert MR, Wick W, Ellingson BM, Hashimoto N, Pollack IF, Brandes AA, Franceschi E, Herold-Mende C, Nayak L, Panigrahy A, Pope WB, Prins R, Sampson JH, Wen PY, Reardon DA. Immunotherapy response assessment in neuro-oncology: a report of the RANO working group. Lancet Oncology. 2015;16(15):534-542. https://doi.org/10.1016/s1470-2045(15)00088-1

Email Confirmation

An email was sent to test@gmail.com with a confirmation link. Follow the link from the letter to complete the registration on the site.

Email Confirmation

We use cооkies to improve the performance of the site. By staying on our site, you agree to the terms of use of cооkies. To view our Privacy and Cookie Policy, please. click here.