1
|
Maghrouni A, Givari M, Jalili-Nik M, Mollazadeh H, Bibak B, Sadeghi MM, Afshari AR, Johnston TP, Sahebkar A. Targeting the PD-1/PD-L1 pathway in glioblastoma multiforme: Preclinical evidence and clinical interventions. Int Immunopharmacol 2021; 93:107403. [PMID: 33581502 DOI: 10.1016/j.intimp.2021.107403] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/11/2020] [Accepted: 12/23/2020] [Indexed: 12/14/2022]
Abstract
Glioblastoma multiforme (GBM), as one of the immunosuppressive and common intrinsic brain tumors in adults, remains an intractable malignancy to manage. Since the standard of care for treatment, which includes surgery and chemoradiation, has not provided a sustainable and durable response in affected patients, seeking novel therapeutic approaches to treat GBM seems imperative. Immunotherapy, a breakthrough for cancer treatment, has become an attractive tool for combating cancer with the potential to access the blood-brain-barrier (BBB). In this regard, programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1), as major immunological checkpoints, have drawn considerable interest due to their effectiveness in a spectrum of highly-aggressive neoplasms through negative regulation of the T-cell-mediated immune response. Nevertheless, due to the immunosuppressive microenvironment of GBM, the efficacy of these immune checkpoint inhibitors (ICIs), when used as monotherapy, has been unfavorable and lacks sufficient beneficial outcomes for GBM patients. A variety of clinical studies are attempting to evaluate the combination of ICIs (neoadjuvant/adjuvant) and existing treatment guidelines to strengthen their effectiveness; however, the exact mechanism of this signaling axis affects the consequences of immune therapy remains elusive. This review provides an overview of the PD-1/PD-L1 pathway, currently approved ICIs for clinical use, preclinical and clinical trials of PD-1/PD-L1 as monotherapy, and when used concomitantly with other GBM treatments.
Collapse
Affiliation(s)
- Abolfazl Maghrouni
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Givari
- Department of Laboratory Sciences, School of Paramedical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Jalili-Nik
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Mollazadeh
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Bahram Bibak
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mohammad Montazami Sadeghi
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Amir R Afshari
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Patti G, Calandra E, De Bellis A, Gallizia A, Crocco M, Napoli F, Allegri AME, Thiabat HF, Bellastella G, Maiorino MI, Garrè ML, Parodi S, Maghnie M, di Iorgi N. Antibodies Against Hypothalamus and Pituitary Gland in Childhood-Onset Brain Tumors and Pituitary Dysfunction. Front Endocrinol (Lausanne) 2020; 11:16. [PMID: 32132974 PMCID: PMC7040196 DOI: 10.3389/fendo.2020.00016] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 01/09/2020] [Indexed: 12/14/2022] Open
Abstract
Purpose: To detect the presence of antipituitary (APA) and antihypothalamus antibodies (AHA) in subjects treated for brain cancers, and to evaluate their potential association with pituitary dysfunction. Methods: We evaluated 63 patients with craniopharyngioma, glioma, and germinoma treated with surgery and/or radiotherapy and/or chemotherapy at a median age of 13 years. Forty-one had multiple pituitary hormone deficiencies (MPHD), six had a single pituitary defect. GH was the most common defect (65.1%), followed by AVP (61.9%), TSH (57.1%), ACTH (49.2%), and gonadotropin (38.1%). APA and AHA were evaluated by simple indirect immunofluorescence method indirect immunofluorescence in patients and in 50 healthy controls. Results: Circulating APA and/or AHA were found in 31 subjects (49.2%) and in none of the healthy controls. In particular, 25 subjects out of 31 were APA (80.6%), 26 were AHA (83.90%), and 20 were both APA and AHA (64.5%). Nine patients APA and/or AHA have craniopharyngioma (29%), seven (22.6%) have glioma, and 15 (48.4%) have germinoma. Patients with craniopharyngioma were positive for at least one antibody in 39.1% compared to 33.3% of patients with glioma and to 78.9% of those with germinoma with an analogous distribution for APA and AHA between the three tumors. The presence of APA or AHA and of both APA and AHA was significantly increased in patients with germinoma. The presence of APA (P = 0.001) and their titers (P = 0.001) was significantly associated with the type of tumor in the following order: germinomas, craniopharyngiomas, and gliomas; an analogous distribution was observed for the presence of AHA (P = 0.002) and their titers (P = 0.012). In addition, we found a significant association between radiotherapy and APA (P = 0.03). Conclusions: Brain tumors especially germinoma are associated with the development of hypothalamic-pituitary antibodies and pituitary defects. The correct interpretation of APA/AHA antibodies is essential to avoid a misdiagnosis of an autoimmune infundibulo-neurohypophysitis or pituitary hypophysitis in patients with germinoma.
Collapse
Affiliation(s)
- Giuseppa Patti
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, University of Genova, Genova, Italy
- Department of Neuroscience, Rehabilitation, Ophtalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Erika Calandra
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, University of Genova, Genova, Italy
- Department of Neuroscience, Rehabilitation, Ophtalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Annamaria De Bellis
- Department of Advanced Medical and Surgical Sciences Endocrinology and Metabolic Unit, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Annalisa Gallizia
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, University of Genova, Genova, Italy
- Department of Neuroscience, Rehabilitation, Ophtalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Marco Crocco
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, University of Genova, Genova, Italy
- Department of Neuroscience, Rehabilitation, Ophtalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Flavia Napoli
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | - Hanan F. Thiabat
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, University of Genova, Genova, Italy
- Department of Neuroscience, Rehabilitation, Ophtalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Giuseppe Bellastella
- Department of Advanced Medical and Surgical Sciences Endocrinology and Metabolic Unit, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Maria Ida Maiorino
- Department of Advanced Medical and Surgical Sciences Endocrinology and Metabolic Unit, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Stefano Parodi
- Epidemiology and Biostatistics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Mohamad Maghnie
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, University of Genova, Genova, Italy
- Department of Neuroscience, Rehabilitation, Ophtalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
- *Correspondence: Mohamad Maghnie ;
| | - Natascia di Iorgi
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, University of Genova, Genova, Italy
- Department of Neuroscience, Rehabilitation, Ophtalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| |
Collapse
|
3
|
Abstract
PURPOSE Given its extremely poor prognosis, there is a pressing need for an improved understanding of the biology of glioblastoma multiforme (GBM), including the roles of tumor subpopulations that may contribute to their growth rate and therapy resistance. The most malignant phenotypes of GBM have been ascribed to the presence of subpopulations of cancer stem cells (CSCs), which are resistant to chemotherapeutic drugs and ionizing radiation and which promote invasiveness and metastasis. The mechanisms by which the CSC state is obtained and by which it promotes tumor maintenance are only beginning to emerge. We hypothesize that M2 polarized macrophages may affect CSC phenotypes via cell-cell communication. METHODS We investigated the interplay between glioma CSCs and macrophages via co-culture. The invasiveness of CSCs in the absence and presence of macrophages was assessed using collagen degradation and Transwell migration assays. The role of STAT3 as a CSC phenotypic mediator was assessed using siRNA-mediated gene silencing. RESULTS We found that the levels of a M2 macrophage-specific secreted cytokine, TGF-β1, were elevated in the presence of CSCs, regardless of whether the cells were plated as contacting or non-contacting co-cultures. In addition, we found that the co-culture resulted in enhanced expression of M2 markers in macrophages that were previously polarized to the M1 phenotype. siRNA-mediated STAT3 silencing was found to reduce the chemo-responsiveness and migratory abilities of the CSCs. Combination treatment of STAT3 siRNA and DNA alkylating agents was found to further abrogate CSC functions. CONCLUSIONS Our data indicate that the co-culture of CSCs and macrophages results in bi-directional signaling that alters the phenotypes of both cell types. These results provide an explanation for recently observed effects of macrophages on GBM tumor cell growth, motility and therapeutic resistance, and suggest potential therapeutic strategies to disrupt the CSC phenotype by impairing its communication with macrophages.
Collapse
Affiliation(s)
- Leora M Nusblat
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Molly J Carroll
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA.,Department of Biomedical Engineering, University of Wisconsin, Madison, WI, 53706, USA
| | - Charles M Roth
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA. .,Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA.
| |
Collapse
|
4
|
Abstract
Glioblastoma multiforme (GBM) is the most common primary brain tumor and is notorious for its poor prognosis. The highly invasive nature of GBM and its inherent resistance to therapy lead to very high rates of recurrence. Recently, a small cohort of tumor cells, called cancer stem cells (CSCs), has been recognized as a subset of tumor cells with self-renewal ability and multilineage capacity. These properties, along with the remarkable tumorigenicity of CSCs, are thought to account for the high rates of tumor recurrence after treatment. Recent research has been geared toward understanding the unique biological characteristics of CSCs to enable development of targeted therapy. Strategies include inhibition of CSC-specific pathways and receptors; agents that increase sensitivity of CSCs to chemotherapy and radiotherapy; CSC differentiation agents; and CSC-specific immunotherapy, virotherapy, and gene therapy. These approaches could inform the development of newer therapeutics for GBM.
Collapse
|
5
|
Immunobiology and immunotherapeutic targeting of glioma stem cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 853:139-66. [PMID: 25895711 DOI: 10.1007/978-3-319-16537-0_8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
For decades human brain tumors have confounded our efforts to effectively manage and treat patients. In adults, glioblastoma multiforme is the most common malignant brain tumor with a patient survival of just over 14 months. In children, brain tumors are the leading cause of solid tumor cancer death and gliomas account for one-fifth of all childhood cancers. Despite advances in conventional treatments such as surgical resection, radiotherapy, and systemic chemotherapy, the incidence and mortality rates for gliomas have essentially stayed the same. Furthermore, research efforts into novel therapeutics that initially appeared promising have yet to show a marked benefit. A shocking and somewhat disturbing view is that investigators and clinicians may have been targeting the wrong cells, resulting in the appearance of the removal or eradication of patient gliomas only to have brain cancer recurrence. Here we review research progress in immunotherapy as it pertains to glioma treatment and how it can and is being adapted to target glioma stem cells (GSCs) as a means of dealing with this potential paradigm.
Collapse
|
6
|
Cao JX, Zhang XY, Liu JL, Li D, Li JL, Liu YS, Wang M, Xu BL, Wang HB, Wang ZX. Clinical efficacy of tumor antigen-pulsed DC treatment for high-grade glioma patients: evidence from a meta-analysis. PLoS One 2014; 9:e107173. [PMID: 25215607 PMCID: PMC4162602 DOI: 10.1371/journal.pone.0107173] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 08/07/2014] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND The effectiveness of immunotherapy for high-grade glioma (HGG) patients remains controversial. To evaluate the therapeutic efficacy of dendritic cells (DCs) alone in the treatment of HGG, we performed a systematic review and meta-analysis in terms of patient survival with relevant published clinical studies. MATERIALS AND METHODS A total of 409 patients, including historical cohorts, nonrandomized and randomized controls with HGG, were selected for the meta-analysis. RESULTS The treatment of HGG with DCs was associated with a significantly improved one-year survival (OS) (p<0.001) and 1.5-, 2-, 3-, 4-, and 5-year OS (p<0.001) compared with the non-DC group. A meta-analysis of the patient outcome data revealed that DC immunotherapy has a significant influence on progression-free survival (PFS) in HGG patients, who showed significantly improved 1-,1.5-, 2-, 3- and 4-year PFS (p<0.001). The analysis of Karnofsky performance status (KPS) demonstrated no favorable results for DC cell therapy arm (p = 0.23).The percentages of CD3+CD8+ and CD3+CD4+ T cells and CD16+ lymphocyte subset were not significantly increased in the DC group compared with the baseline levels observed before treatment (p>0.05), whereas CD56+ lymphocyte subset were significantly increased after DC treatment (p = 0.0001). Furthermore, the levels of IFN-γ in the peripheral blood of HGG patients, which reflect the immune function of the patients, were significantly increased after DC immunotherapy (p<0.001). CONCLUSIONS Thus, our meta-analysis showed that DC immunotherapy markedly prolongs survival rates and progression-free time, enhances immune function, and improves the efficacy of the treatment of HGG patients.
Collapse
Affiliation(s)
- Jun-Xia Cao
- Biotherapy Center, the General Hospital of Beijing Military Command, Beijing, People's Republic of China
- Tsinghua-Peking Center for Life Sciences, Laboratory of Dynamic Immunobiology, School of Medicine, School of Life Sciences, Tsinghua University, Beijing, People's Republic of China
- * E-mail: (ZXW); (JXC)
| | - Xiao-Yan Zhang
- Biotherapy Center, the General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Jin-Long Liu
- Biotherapy Center, the General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Duo Li
- Biotherapy Center, the General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Jun-Li Li
- Biotherapy Center, the General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Yi-Shan Liu
- Biotherapy Center, the General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Min Wang
- Biotherapy Center, the General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Bei-Lei Xu
- Biotherapy Center, the General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Hai-Bo Wang
- Biotherapy Center, the General Hospital of Beijing Military Command, Beijing, People's Republic of China
| | - Zheng-Xu Wang
- Biotherapy Center, the General Hospital of Beijing Military Command, Beijing, People's Republic of China
- * E-mail: (ZXW); (JXC)
| |
Collapse
|
7
|
Glioma-associated antigen HEATR1 induces functional cytotoxic T lymphocytes in patients with glioma. J Immunol Res 2014; 2014:131494. [PMID: 25126583 PMCID: PMC4121097 DOI: 10.1155/2014/131494] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Revised: 05/17/2014] [Accepted: 06/16/2014] [Indexed: 11/18/2022] Open
Abstract
A2B5+ glioblastoma (GBM) cells have glioma stem-like cell (GSC) properties that are crucial to chemotherapy resistance and GBM relapse. T-cell-based antigens derived from A2B5+ GBM cells provide important information for immunotherapy. Here, we show that HEAT repeat containing 1 (HEATR1) expression in GBM tissues was significantly higher than that in control brain tissues. Furthermore, HEATR1 expression in A2B5+ U87 cells was higher than that in A2B5-U87 cells (P = 0.016). Six peptides of HEATR1 presented by HLA-A∗02 were selected for testing of their ability to induce T-cell responses in patients with GBM. When peripheral blood mononuclear cells from healthy donors (n = 6) and patients with glioma (n = 33) were stimulated with the peptide mixture, eight patients with malignant gliomas had positive reactivity with a significantly increased number of responding T-cells. The peptides HEATR(1682-690), HEATR(11126-1134), and HEATR(1757-765) had high affinity for binding to HLA-A∗02:01 and a strong capacity to induce CTL response. CTLs against HEATR1 peptides were capable of recognizing and lysing GBM cells and GSCs. These data are the first to demonstrate that HEATR1 could induce specific CTL responses targeting both GBM cells and GSCs, implicating that HEATR1 peptide-based immunotherapy could be a novel promising strategy for treating patients with GBM.
Collapse
|
8
|
Xia H, Yu C, Zhang W, Zhang B, Zhao Y, Fang F. [Identification and isolation of cancer stem cells from A549 cells]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2014; 16:400-4. [PMID: 23945242 PMCID: PMC6000669 DOI: 10.3779/j.issn.1009-3419.2013.08.02] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
背景与目的 肺癌干细胞是肺癌恶性表型的根源和潜在的治疗靶点,从人肺癌A549细胞株中分离肺癌干细胞,观察特异性干细胞标志物分子的表达,为进一步的干细胞研究提供试验基础。 方法 接种肺癌A549细胞株,经流式细胞术,特异性筛选分离肺癌干细胞,观察克隆形成能力、细胞增殖能力和体外致瘤能力的差别,并分别用RT-PCR和Western blot的方法分析干细胞标志物分子CD133和ABCG2的表达。 结果 经过流式细胞仪成功分选了人肺腺癌A549细胞系SP细胞亚群,结果表明此SP细胞亚群约占A549细胞总数的5.93%,经维拉帕米处理后Hoechest33342阴性/弱阳性细胞含量下降为0.32%,SP细胞克隆形成能力,细胞增殖能力和体外致瘤能力均明显高于非SP细胞。RT-PCR和Western blot结果发现,筛选分离的肺癌SP细胞群高表达干细胞标志物分子CD133和ABCG2。 结论 通过流式细胞术可以筛选分离高表达CD133和ABCG2分子的肺癌干细胞,可用于进一步的研究中。
Collapse
Affiliation(s)
- Hui Xia
- Department of Carciothoracic Surgery, the First Affiliated Hospital of the General Hospital of PLA, Beijing 100048, China.
| | | | | | | | | | | |
Collapse
|
9
|
Ma C, Li Y, Zhang X, Zhao G, Xu H. Levels of vascular endothelial growth factor and matrix metalloproteinase-9 proteins in patients with glioma. J Int Med Res 2014; 42:198-204. [PMID: 24398760 DOI: 10.1177/0300060513481924] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To investigate the levels of vascular endothelial growth factor (VEGF) and matrix metalloproteinase-9 (MMP-9) proteins in patients with glioma, in order to determine if either protein has prognostic value. METHOD The presence of VEGF and MMP-9 proteins in paraffin-embedded tumour specimens from patients with glioma was detected using immunohistochemistry. The correlation between the levels of VEGF and MMP-9 proteins and tumour grade was analysed. RESULTS A total of 32 patients with low-grade gliomas (World Health Organization [WHO] grade II) and 48 patients with high-grade gliomas (WHO grades III-IV) participated in the study. Positive immunohistochemical staining of VEGF and MMP-9 proteins was detected in 58/80 (72.5%) and 60/80 (75.0%) of patients, respectively. The level of VEGF immunostaining was significantly positively correlated with the level of MMP-9 immunostaining (r = 0.78). Significantly more high-grade gliomas (grades III-IV) demonstrated positive VEGF and MMP-9 immunostaining compared with the low grade gliomas (grades I-II). CONCLUSIONS These data suggest that VEGF and MMP-9 play an important role in the malignant behaviour of gliomas.
Collapse
Affiliation(s)
- Chengyuan Ma
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, Jilin Province, China
| | | | | | | | | |
Collapse
|
10
|
Glioma stem cells and immunotherapy for the treatment of malignant gliomas. ISRN ONCOLOGY 2013; 2013:673793. [PMID: 23762610 PMCID: PMC3671309 DOI: 10.1155/2013/673793] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/03/2013] [Accepted: 03/27/2013] [Indexed: 02/06/2023]
Abstract
Stem cell research has led to the discovery of glioma stem cells (GSCs), and because these cells are resistant to chemotherapy and radiotherapy, analysis of their properties has been rapidly pursued for targeted treatment of malignant glioma. Recent studies have also revealed complex crosstalk between GSCs and their specialized environment (niche). Therefore, targeting not only GSCs but also their niche may be a principle for novel therapies of malignant glioma. One possible novel strategy for targeting GSCs and their niches is immunotherapy with different antitumor mechanism(s) from those of conventional therapy. Recent clinical studies of immunotherapy using peptide vaccines and antibodies have shown promising results. This review describes the recent findings related to GSCs and their niches, as well as immunotherapies for glioma, followed by discussion of immunotherapies that target GSCs for the treatment of malignant glioma.
Collapse
|
11
|
Petrosiute A, Auletta JJ, Lazarus HM. Achieving graft-versus-tumor effect in brain tumor patients: from autologous progenitor cell transplant to active immunotherapy. Immunotherapy 2013. [PMID: 23194364 DOI: 10.2217/imt.12.96] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Success in treating aggressive brain tumors like glioblastoma multiforme and medulloblastoma remains challenging, in part because these malignancies overcome CNS immune surveillance. New insights into brain tumor immunology have led to a rational development of immunotherapeutic strategies, including cytotoxic Tlymphocyte therapies and dendritic cell vaccines. However, these therapies are most effective when applied in a setting of minimal residual disease, so require prior use of standard cytotoxic therapies or cytoreduction by surgery. Myeloablative chemotherapy with autologous hematopoietic cell transplantation (autoHCT) can offer a platform upon which different cellular therapies can be effectively instituted. Specifically, this approach provides an inherent 'chemical debulking' through high-dose chemotherapy and a graft-versus-tumor effect through an autologous T-cell replete graft. Furthermore, autoHCT may be beneficial in 'resetting' the body's immune system, potentially 'breaking' tumor tolerance, and in providing a 'boost' of immune effector cells (NK cells or cytotoxic T lymphocytes), which could augment desired anti-tumor effects. As literature on the use of autoHCT in brain tumors is scarce, aspects of immunotherapies applied in non-CNS malignancies are reviewed as potential therapies that could be used in conjunction with autoHCT to eradicate brain tumors.
Collapse
Affiliation(s)
- Agne Petrosiute
- Department of Pediatrics, Hematology/Oncology, Rainbow Babies & Children's Hospital, Case Western Reserve University, 11100 Euclid Avenue, Mailstop 6054, Cleveland, OH 44106, USA.
| | | | | |
Collapse
|