1
|
Naffaa MM, Al-Ewaidat OA, Gogia S, Begiashvili V. Neoantigen-based immunotherapy: advancing precision medicine in cancer and glioblastoma treatment through discovery and innovation. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002313. [PMID: 40309350 PMCID: PMC12040680 DOI: 10.37349/etat.2025.1002313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025] Open
Abstract
Neoantigen-based immunotherapy has emerged as a transformative approach in cancer treatment, offering precision medicine strategies that target tumor-specific antigens derived from genetic, transcriptomic, and proteomic alterations unique to cancer cells. These neoantigens serve as highly specific targets for personalized therapies, promising more effective and tailored treatments. The aim of this article is to explore the advances in neoantigen-based therapies, highlighting successful treatments such as vaccines, tumor-infiltrating lymphocyte (TIL) therapy, T-cell receptor-engineered T cells therapy (TCR-T), and chimeric antigen receptor T cells therapy (CAR-T), particularly in cancer types like glioblastoma (GBM). Advances in technologies such as next-generation sequencing, RNA-based platforms, and CRISPR gene editing have accelerated the identification and validation of neoantigens, moving them closer to clinical application. Despite promising results, challenges such as tumor heterogeneity, immune evasion, and resistance mechanisms persist. The integration of AI-driven tools and multi-omic data has refined neoantigen discovery, while combination therapies are being developed to address issues like immune suppression and scalability. Additionally, the article discusses the ongoing development of personalized immunotherapies targeting tumor mutations, emphasizing the need for continued collaboration between computational and experimental approaches. Ultimately, the integration of cutting-edge technologies in neoantigen research holds the potential to revolutionize cancer care, offering hope for more effective and targeted treatments.
Collapse
Affiliation(s)
- Moawiah M Naffaa
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ola A Al-Ewaidat
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL 60202, USA
| | - Sopiko Gogia
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL 60202, USA
| | - Valiko Begiashvili
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66103, USA
| |
Collapse
|
2
|
Pour ME, Moghadam SG, Shirkhani P, Sahebkar A, Mosaffa F. Therapeutic cell-based vaccines for glioblastoma multiforme. Med Oncol 2023; 40:354. [PMID: 37952224 DOI: 10.1007/s12032-023-02220-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 10/12/2023] [Indexed: 11/14/2023]
Abstract
Glioblastoma multiforme (GBM), a highly aggressive tumor, poses significant challenges in achieving successful treatment outcomes. Conventional therapeutic modalities including surgery, radiation, and chemotherapy have demonstrated limited efficacy, primarily attributed to the complexities associated with drug delivery to the tumor site and tumor heterogeneity. To address this critical need for innovative therapies, the potential of cancer vaccines utilizing tumor cells and dendritic cells has been explored for GBM treatment. This article provides a comprehensive review of therapeutic vaccinations employing cell-based vaccine strategies for the management of GBM. A meticulous evaluation of 45 clinical trials involving more than 1500 participants revealed that cell-based vaccinations have exhibited favorable safety profiles with minimal toxicity. Moreover, these vaccines have demonstrated modest improvements in overall survival and progression-free survival among patients. However, certain limitations still persist. Notably, there is a need for advancements in the development of potent antigens to evoke immune responses, as well as the optimization of dosage regimens. Consequently, while cell-based vaccinations show promise as a potential therapeutic approach for GBM, further research is imperative to overcome the current limitations. The ultimate objective is to surmount these obstacles and establish cell-based vaccinations as a standard therapeutic modality for GBM.
Collapse
Affiliation(s)
- Mehrshad Ebrahim Pour
- School of Pharmacy, Department of Pharmaceutical Biotechnology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samin Ghorbani Moghadam
- School of Pharmacy, Department of Pharmaceutical Biotechnology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parian Shirkhani
- School of Pharmacy, Department of Pharmaceutical Biotechnology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Mosaffa
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Baddam SR, Kalagara S, Kuna K, Enaganti S. Recent advancements and theranostics strategies in glioblastoma therapy. Biomed Mater 2023; 18:052007. [PMID: 37582381 DOI: 10.1088/1748-605x/acf0ab] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 08/15/2023] [Indexed: 08/17/2023]
Abstract
Glioblastoma (GBM) is the most aggressive and lethal malignant brain tumor, and it is challenging to cure with surgery and treatment. The prevention of permanent brain damage and tumor invasion, which is the ultimate cause of recurrence, are major obstacles in GBM treatment. Besides, emerging treatment modalities and newer genetic findings are helping to understand and manage GBM in patients. Accordingly, researchers are focusing on advanced nanomaterials-based strategies for tackling the various problems associated with GBM. In this context, researchers explored novel strategies with various alternative treatment approaches such as early detection techniques and theranostics approaches. In this review, we have emphasized the recent advancement of GBM cellular models and their roles in designing GBM therapeutics. We have added a special emphasis on the novel genetic and drug target findings as well as strategies for early detection. Besides, we have discussed various theranostic approaches such as hyperthermia therapy, phototherapy and image-guided therapy. Approaches utilized for targeted drug delivery to the GBM were also discussed. This article also describes the recentin vivo, in vitroandex vivoadvances using innovative theranostic approaches.
Collapse
Affiliation(s)
- Sudhakar Reddy Baddam
- University of Massachusetts Chan Medical School, RNA Therapeutics Institute,Worcester,MA 01655, United States of America
| | - Sudhakar Kalagara
- Department of Chemistry and Biochemistry,University of the Texas at El Paso, 500 W University Ave,El Paso,TX 79968, United States of America
| | - Krishna Kuna
- Department of Chemistry,University College of Science, Saifabad, Osmania University, Hyderabad,Telangana,India
| | - Sreenivas Enaganti
- Department of Bioinformatics, Averinbiotech Laboratories,208, 2nd Floor, Windsor Plaza, Nallakunta, Hyderabad, Telangana,India
| |
Collapse
|
4
|
Das S, Dash BS, Premji TP, Chen JP. Immunotherapeutic Approaches for the Treatment of Glioblastoma Multiforme: Mechanism and Clinical Applications. Int J Mol Sci 2023; 24:10546. [PMID: 37445721 PMCID: PMC10341481 DOI: 10.3390/ijms241310546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Glioma is one of the most aggressive types of primary brain tumor with a high-grade glioma known as glioblastoma multiforme (GBM). Patients diagnosed with GBM usually have an overall survival rate of less than 18 months after conventional therapy. This bleak prognosis underlines the need to consider new therapeutic interventions for GBM treatment to overcome current treatment limitations. By highlighting different immunotherapeutic approaches currently in preclinical and clinical trials, including immune checkpoint inhibitors, chimeric antigen receptors T cells, natural killer cells, vaccines, and combination therapy, this review aims to discuss the mechanisms, benefits, and limitations of immunotherapy in treating GBM patients.
Collapse
Affiliation(s)
- Suprava Das
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; (S.D.); (B.S.D.); (T.P.P.)
| | - Banendu Sunder Dash
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; (S.D.); (B.S.D.); (T.P.P.)
| | - Thejas P. Premji
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; (S.D.); (B.S.D.); (T.P.P.)
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan; (S.D.); (B.S.D.); (T.P.P.)
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Kwei-San, Taoyuan 33305, Taiwan
- Craniofacial Research Center, Chang Gung Memorial Hospital at Linkou, Kwei-San, Taoyuan 33305, Taiwan
- Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Kwei-San, Taoyuan 33305, Taiwan
- Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan
| |
Collapse
|
5
|
Fisher JP, Adamson DC. Current FDA-Approved Therapies for High-Grade Malignant Gliomas. Biomedicines 2021; 9:biomedicines9030324. [PMID: 33810154 PMCID: PMC8004675 DOI: 10.3390/biomedicines9030324] [Citation(s) in RCA: 175] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 12/26/2022] Open
Abstract
The standard of care (SOC) for high-grade gliomas (HGG) is maximally safe surgical resection, followed by concurrent radiation therapy (RT) and temozolomide (TMZ) for 6 weeks, then adjuvant TMZ for 6 months. Before this SOC was established, glioblastoma (GBM) patients typically lived for less than one year after diagnosis, and no adjuvant chemotherapy had demonstrated significant survival benefits compared with radiation alone. In 2005, the Stupp et al. randomized controlled trial (RCT) on newly diagnosed GBM patients concluded that RT plus TMZ compared to RT alone significantly improved overall survival (OS) (14.6 vs. 12.1 months) and progression-free survival (PFS) at 6 months (PFS6) (53.9% vs. 36.4%). Outside of TMZ, there are four drugs and one device FDA-approved for the treatment of HGGs: lomustine, intravenous carmustine, carmustine wafer implants, bevacizumab (BVZ), and tumor treatment fields (TTFields). These treatments are now mainly used to treat recurrent HGGs and symptoms. TTFields is the only treatment that has been shown to improve OS (20.5 vs. 15.6 months) and PFS6 (56% vs. 37%) in comparison to the current SOC. TTFields is the newest addition to this list of FDA-approved treatments, but has not been universally accepted yet as part of SOC.
Collapse
Affiliation(s)
- Jacob P. Fisher
- Division of Biochemistry, Southern Virginia University, Buena Vista, VA 24416, USA
- Correspondence:
| | - David C. Adamson
- Department of Neurosurgery, School of Medicine, Emory University, Atlanta, GA 30322, USA;
- Atlanta VA Healthcare System, Decatur, GA 30033, USA
| |
Collapse
|
6
|
Davidson TB, Lee A, Hsu M, Sedighim S, Orpilla J, Treger J, Mastall M, Roesch S, Rapp C, Galvez M, Mochizuki A, Antonios J, Garcia A, Kotecha N, Bayless N, Nathanson D, Wang A, Everson R, Yong WH, Cloughesy TF, Liau LM, Herold-Mende C, Prins RM. Expression of PD-1 by T Cells in Malignant Glioma Patients Reflects Exhaustion and Activation. Clin Cancer Res 2018; 25:1913-1922. [PMID: 30498094 DOI: 10.1158/1078-0432.ccr-18-1176] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/27/2018] [Accepted: 11/26/2018] [Indexed: 12/16/2022]
Abstract
PURPOSE Glioblastoma (GBM) is the most common primary malignant tumor in the central nervous system. Our recent preclinical work has suggested that PD-1/PD-L1 plays an important immunoregulatory role to limit effective antitumor T-cell responses induced by active immunotherapy. However, little is known about the functional role that PD-1 plays on human T lymphocytes in patients with malignant glioma.Experimental Design: In this study, we examined the immune landscape and function of PD-1 expression by T cells from tumor and peripheral blood in patients with malignant glioma. RESULTS We found several differences between PD-1+ tumor-infiltrating lymphocytes (TIL) and patient-matched PD-1+ peripheral blood T lymphocytes. Phenotypically, PD-1+ TILs exhibited higher expression of markers of activation and exhaustion than peripheral blood PD-1+ T cells, which instead had increased markers of memory. A comparison of the T-cell receptor variable chain populations revealed decreased diversity in T cells that expressed PD-1, regardless of the location obtained. Functionally, peripheral blood PD-1+ T cells had a significantly increased proliferative capacity upon activation compared with PD-1- T cells. CONCLUSIONS Our evidence suggests that PD-1 expression in patients with glioma reflects chronically activated effector T cells that display hallmarks of memory and exhaustion depending on its anatomic location. The decreased diversity in PD-1+ T cells suggests that the PD-1-expressing population has a narrower range of cognate antigen targets compared with the PD-1 nonexpression population. This information can be used to inform how we interpret immune responses to PD-1-blocking therapies or other immunotherapies.
Collapse
Affiliation(s)
- Tom B Davidson
- Department of Pediatrics, Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California.,Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
| | - Alexander Lee
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California.,Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
| | - Melody Hsu
- Department of Pediatrics, Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
| | - Shaina Sedighim
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
| | - Joey Orpilla
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
| | - Janet Treger
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
| | - Max Mastall
- Division of Experimental Neurosurgery, Department of Neurosurgery, University of Heidelberg, Heidelberg, Germany
| | - Saskia Roesch
- Division of Experimental Neurosurgery, Department of Neurosurgery, University of Heidelberg, Heidelberg, Germany
| | - Carmen Rapp
- Division of Experimental Neurosurgery, Department of Neurosurgery, University of Heidelberg, Heidelberg, Germany
| | - Mildred Galvez
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
| | - Aaron Mochizuki
- Department of Pediatrics, Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
| | - Joseph Antonios
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
| | - Alejandro Garcia
- Department of Medicine/Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
| | - Nikesh Kotecha
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Nicholas Bayless
- Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - David Nathanson
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
| | - Anthony Wang
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
| | - Richard Everson
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
| | - William H Yong
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
| | - Timothy F Cloughesy
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California.,Department of Neurology, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
| | - Linda M Liau
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California.,Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California.,Brain Research Institute, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
| | - Christel Herold-Mende
- Division of Experimental Neurosurgery, Department of Neurosurgery, University of Heidelberg, Heidelberg, Germany
| | - Robert M Prins
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California. .,Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California.,Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California.,Parker Institute for Cancer Immunotherapy, San Francisco, California.,Brain Research Institute, David Geffen School of Medicine at UCLA, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
7
|
Antonios JP, Soto H, Everson RG, Moughon D, Orpilla JR, Shin NP, Sedighim S, Treger J, Odesa S, Tucker A, Yong WH, Li G, Cloughesy TF, Liau LM, Prins RM. Immunosuppressive tumor-infiltrating myeloid cells mediate adaptive immune resistance via a PD-1/PD-L1 mechanism in glioblastoma. Neuro Oncol 2018; 19:796-807. [PMID: 28115578 DOI: 10.1093/neuonc/now287] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background Adaptive immune resistance in the tumor microenvironment appears to attenuate the immunotherapeutic targeting of glioblastoma (GBM). In this study, we identified a tumor-infiltrating myeloid cell (TIM) population that expands in response to dendritic cell (DC) vaccine treatment. The aim of this study was to understand how this programmed death ligand 1 (PD-L1)-expressing population restricts activation and tumor-cytolytic function of vaccine-induced tumor-infiltrating lymphocytes (TILs). Methods To test this hypothesis in our in vivo preclinical model, we treated mice bearing intracranial gliomas with DC vaccination ± murine anti-PD-1 monoclonal antibody (mAb) blockade or a colony stimulating factor 1 receptor inhibitor (CSF-1Ri) (PLX3397) and measured overall survival. We then harvested and characterized the PD-L1+ TIM population and its role in TIL activation and tumor cytolysis in vitro. Results Our data indicated that the majority of PD-L1 expression in the GBM environment is contributed by TIMs rather than by tumor cells themselves. While PD-1 blockade partially reversed the TIL dysfunction, targeting TIMs directly with CSF-1Ri altered TIM expression of key chemotactic factors associated with promoting increased TIL infiltration after vaccination. Neither PD-1 mAb nor CSF-1Ri had a demonstrable therapeutic benefit alone, but when combined with DC vaccination, a significant survival benefit was observed. When the tripartite regimen was given (DC vaccine, PD-1 mAb, PLX3397), long-term survival was noted together with an increase in the number of TILs and TIL activation. Conclusion Together, these studies elucidate the role that TIMs play in mediating adaptive immune resistance in the GBM microenvironment and provide evidence that they can be manipulated pharmacologically with agents that are clinically available. Development of immune resistance in response to active vaccination in GBM can be reversed with dual administration of CSF-1Ri and PD-1 mAb.
Collapse
Affiliation(s)
- Joseph P Antonios
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA
| | - Horacio Soto
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA
| | - Richard G Everson
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA
| | - Diana Moughon
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Joey R Orpilla
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA
| | - Namjo P Shin
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA
| | - Shaina Sedighim
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA
| | - Janet Treger
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA
| | - Sylvia Odesa
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA
| | - Alexander Tucker
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA
| | - William H Yong
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA
| | - Gang Li
- Department of Biostatistics, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA
| | - Timothy F Cloughesy
- Brain Research Institute, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA.,Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA.,Department of Neurology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA
| | - Linda M Liau
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA.,Brain Research Institute, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA.,Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA
| | - Robert M Prins
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA.,Brain Research Institute, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA.,Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA.,Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA
| |
Collapse
|
8
|
Antonios JP, Soto H, Everson RG, Orpilla J, Moughon D, Shin N, Sedighim S, Yong WH, Li G, Cloughesy TF, Liau LM, Prins RM. PD-1 blockade enhances the vaccination-induced immune response in glioma. JCI Insight 2016; 1. [PMID: 27453950 DOI: 10.1172/jci.insight.87059] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
DC vaccination with autologous tumor lysate has demonstrated promising results for the treatment of glioblastoma (GBM) in preclinical and clinical studies. While the vaccine appears capable of inducing T cell infiltration into tumors, the effectiveness of active vaccination in progressively growing tumors is less profound. In parallel, a number of studies have identified negative costimulatory pathways, such as programmed death 1/programmed death ligand 1 (PD-1/PD-L1), as relevant mediators of the intratumoral immune responses. Clinical responses to PD-1 pathway inhibition, however, have also been varied. To evaluate the relevance to established glioma, the effects of PD-1 blockade following DC vaccination were tested in intracranial (i.c.) glioma tumor- bearing mice. Treatment with both DC vaccination and PD-1 mAb blockade resulted in long-term survival, while neither agent alone induced a survival benefit in animals with larger, established tumors. This survival benefit was completely dependent on CD8+ T cells. Additionally, DC vaccine plus PD-1 mAb blockade resulted in the upregulation of integrin homing and immunologic memory markers on tumor-infiltrating lymphocytes (TILs). In clinical samples, DC vaccination in GBM patients was associated with upregulation of PD-1 expression in vivo, while ex vivo blockade of PD-1 on freshly isolated TILs dramatically enhanced autologous tumor cell cytolysis. These findings strongly suggest that the PD-1/PD-L1 pathway plays an important role in the adaptive immune resistance of established GBM in response to antitumor active vaccination and provide us with a rationale for the clinical translation of this combination therapy.
Collapse
Affiliation(s)
- Joseph P Antonios
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA
| | - Horacio Soto
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA
| | - Richard G Everson
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA
| | - Joey Orpilla
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA
| | - Diana Moughon
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA
| | - Namjo Shin
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA
| | - Shaina Sedighim
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA
| | - William H Yong
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA
| | - Gang Li
- Department of Biostatistics, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA
| | - Timothy F Cloughesy
- Brain Research Institute, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA; Jonsson Comprehensive Cancer Center (JCCC), David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA; Department of Neurology, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA
| | - Linda M Liau
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA; Brain Research Institute, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA; Jonsson Comprehensive Cancer Center (JCCC), David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA
| | - Robert M Prins
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA; Brain Research Institute, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA; Jonsson Comprehensive Cancer Center (JCCC), David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, UCLA, Los Angeles, California, USA
| |
Collapse
|
9
|
Dong S, Xu T, Zhao P, Parent KN, Chen M. A Comparison Study of iTEP Nanoparticle-Based CTL Vaccine Carriers Revealed a Surprise Relationship between the Stability and Efficiency of the Carriers. Am J Cancer Res 2016; 6:666-78. [PMID: 27022414 PMCID: PMC4805661 DOI: 10.7150/thno.14068] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 02/08/2016] [Indexed: 01/08/2023] Open
Abstract
Vaccine carriers have been shown to enhance cytotoxic T lymphocyte (CTL) epitope peptide vaccines by addressing intrinsic limitations of the vaccines. We have previously developed an immune-tolerant elastin-like polypeptide (iTEP)-based nanoparticle (NP) as an effective and unique CTL vaccine carrier. The NP is unique for its humoral immune tolerance, flexible structure, and ability to deliver CTL vaccines as polypeptide fusions. Here, we aimed to improve the NP by increasing its stability since we found it was not stable. We thus generated a more stable iTEP NP (ST-NP) and used it to deliver a CTL peptide vaccine, SIINFEKL. However, we surprisingly found that the ST-NP had a lower efficiency than the previously developed, marginally stable iTEP NP (MS-NP) in terms of promoting vaccine presentation and vaccine-induced CTL responses. On the other hand, dendritic cells (DCs) showed preferential uptake of the ST-NP but not the MS-NP. To develop an iTEP vaccine carrier that outperforms both the MS-NP and the ST-NP, we devised an iTEP NP that has a changeable stability responsive to a cytosolic, reductive environment, termed reductive environment-dependent NP or RED-NP. The RED-NP showed an intermediate ability to promote vaccine presentation and T cell responses in vitro between the MS-NP and the ST-NP. However, the RED-NP induced the strongest CTL responses in vivo among all three NPs. In conclusion, iTEP NPs that have a dynamically changeable stability are most effective to deliver and enhance CTL peptide vaccines. The work also demonstrated the versatile nature of iTEP vaccine carriers.
Collapse
|
10
|
Navari M, Zare M, Javanmardi M, Asadi-Ghalehni M, Modjtahedi H, Rasaee MJ. Epitope mapping of epidermal growth factor receptor (EGFR) monoclonal antibody and induction of growth-inhibitory polyclonal antibodies by vaccination with EGFR mimotope. Immunopharmacol Immunotoxicol 2014; 36:309-15. [PMID: 25070131 DOI: 10.3109/08923973.2014.945127] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
One of the proposed approaches in cancer therapy is to induce and direct the patient's own immune system against cancer cells. In this study, we determined the epitope mapping of the rat anti-human epidermal growth factor receptor (EGFR) monoclonal antibody ICR-62 using a phage display of random peptide library and identified a 12 amino acids peptide, which was recognized as a mimotope. The peptide was synthesized and conjugated to bovine serum albumin (BSA) as carrier protein (P-BSA). We have shown that ICR-62 can react specifically with P-BSA as well as native EGFR. Two rabbits were immunized either by BSA or P-BSA and the rabbits IgGs were purified and examined for binding to the antigens, mimotope and the EGFR protein purified from the EGFR overexpressing A431 cell line. We showed that the rabbit IgG generated against the mimotope is capable of inhibiting the growth of A431 cells by 15%, but does not have any effect on the growth of EGFR-negative MDA-MB-453 cell line in vitro. Our results support the need for further investigations on the potential of vaccination with either mimotope of the EGFR or epitope displayed on the surface of phage particles for use in active immunotherapy of cancer.
Collapse
Affiliation(s)
- Mohsen Navari
- Department of Medical Biotechnology, School of Medical Sciences, Tarbiat Modares University , Tehran , Islamic Republic of Iran and
| | | | | | | | | | | |
Collapse
|
11
|
Addeo R, Zappavigna S, Parlato C, Caraglia M. Erlotinib: early clinical development in brain cancer. Expert Opin Investig Drugs 2014; 23:1027-37. [PMID: 24836441 DOI: 10.1517/13543784.2014.918950] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Glioblastoma (GBM) is the most common brain cancer in adults. It is also, unfortunately, the most aggressive type and the least responsive to therapy. Overexpression of EGFR and/or EGFRvIII is frequently found in GBM and is frequently associated with the more malignant phenotype of the disease and a poor clinical outcome. EGFR-targeted therapy represents a promising anti-GBM therapy. Two EGFR kinase inhibitors, gefitinib and erlotinib have been tested in clinical trials for malignant gliomas. However, the clinical efficacy of EGFR-targeted therapy has been only modest in GBM patients. AREAS COVERED The authors provide an evaluation of erlotinib as a potential therapy for GBM. The authors highlight experiences drawn from clinical trials and discuss the challenges, which include the insufficient penetration through the blood-brain barrier (BBB) and chemoresistance. EXPERT OPINION Malignant brain tumours have a very complex signalling network that is not only driven by EGFR. This complexity dictates tumour sensitivity to EGFR-targeted therapies. Alternative kinase signalling pathways may be involved in parallel with the inhibited target, so that a single target's inactivation is not sufficient to block downstream oncogenic signalling. The use of nanocarriers offers many opportunities, such as the release of the drug to specific cells or tissues, together with the ability to overcome different biological barriers, like the BBB.
Collapse
|
12
|
Wilson TA, Karajannis MA, Harter DH. Glioblastoma multiforme: State of the art and future therapeutics. Surg Neurol Int 2014; 5:64. [PMID: 24991467 PMCID: PMC4078454 DOI: 10.4103/2152-7806.132138] [Citation(s) in RCA: 187] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Accepted: 03/13/2014] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is the most common and lethal primary malignancy of the central nervous system (CNS). Despite the proven benefit of surgical resection and aggressive treatment with chemo- and radiotherapy, the prognosis remains very poor. Recent advances of our understanding of the biology and pathophysiology of GBM have allowed the development of a wide array of novel therapeutic approaches, which have been developed. These novel approaches include molecularly targeted therapies, immunotherapies, and gene therapy. METHODS We offer a brief review of the current standard of care, and a survey of novel therapeutic approaches for treatment of GBM. RESULTS Despite promising results in preclinical trials, many of these therapies have demonstrated limited therapeutic efficacy in human clinical trials. Thus, although survival of patients with GBM continues to slowly improve, treatment of GBM remains extremely challenging. CONCLUSION Continued research and development of targeted therapies, based on a detailed understanding of molecular pathogenesis can reasonably be expected to yield improved outcomes for patients with GBM.
Collapse
Affiliation(s)
- Taylor A Wilson
- Department of Neurosurgery, Division of Oncology, New York University School of Medicine, NY, USA
| | - Matthias A Karajannis
- Department of Pediatrics, Division of Oncology, New York University School of Medicine, NY, USA
| | - David H Harter
- Department of Neurosurgery, Division of Oncology, New York University School of Medicine, NY, USA
| |
Collapse
|
13
|
Schuessler A, Smith C, Beagley L, Boyle GM, Rehan S, Matthews K, Jones L, Crough T, Dasari V, Klein K, Smalley A, Alexander H, Walker DG, Khanna R. Autologous T-cell therapy for cytomegalovirus as a consolidative treatment for recurrent glioblastoma. Cancer Res 2014; 74:3466-76. [PMID: 24795429 DOI: 10.1158/0008-5472.can-14-0296] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Glioblastoma multiforme (GBM) is one of the most aggressive human brain malignancies. Even with optimal treatment, median survival is less than 6 months for patients with recurrent GBM. Immune-based therapies have the potential to improve patient outcome by supplementing standard treatment. Expression of human cytomegalovirus (CMV) antigens in GBM tissues provides the unique opportunity to target viral antigens for GBM therapy. Here, we report findings of a formal clinical assessment of safety and potential clinical efficacy of autologous CMV-specific T-cell therapy as a consolidative treatment for recurrent GBM. From a total of 19 patients with recurrent GBM, CMV-specific T cells were successfully expanded from 13 patients (68.4%), 11 of whom received up to four T-cell infusions. Combination therapy based on T-cell infusion and chemotherapy was well tolerated, and we detected only minor adverse events. The overall survival of these patients since first recurrence ranged from 133 to 2,428 days, with a median overall survival of 403 days. Most importantly, 4 of 10 patients that completed the treatment remained progression free during the study period. Furthermore, molecular profiling of CMV-specific T-cell therapy from these patients revealed distinct gene expression signatures, which correlated with their clinical response. Our study suggests that a combination therapy with autologous CMV-specific T cells and chemotherapy is a safe novel treatment option and may offer clinical benefit for patients with recurrent GBM.
Collapse
Affiliation(s)
- Andrea Schuessler
- Authors' Affiliations: QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory
| | - Corey Smith
- Authors' Affiliations: QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory
| | - Leone Beagley
- Authors' Affiliations: QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory
| | | | - Sweera Rehan
- Authors' Affiliations: QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory
| | - Katherine Matthews
- Authors' Affiliations: QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory
| | - Linda Jones
- Authors' Affiliations: QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory
| | - Tania Crough
- Authors' Affiliations: QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory
| | - Vijayendra Dasari
- Authors' Affiliations: QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory
| | | | - Amy Smalley
- Newro Foundation, The Wesley Hospital, Brisbane, Queensland, Australia
| | - Hamish Alexander
- Authors' Affiliations: QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory; Newro Foundation, The Wesley Hospital, Brisbane, Queensland, Australia
| | - David G Walker
- Newro Foundation, The Wesley Hospital, Brisbane, Queensland, Australia
| | - Rajiv Khanna
- Authors' Affiliations: QIMR Centre for Immunotherapy and Vaccine Development and Tumour Immunology Laboratory;
| |
Collapse
|
14
|
Barnard Z, Wakimoto H, Zaupa C, Patel AP, Klehm J, Martuza RL, Rabkin SD, Curry WT. Expression of FMS-like tyrosine kinase 3 ligand by oncolytic herpes simplex virus type I prolongs survival in mice bearing established syngeneic intracranial malignant glioma. Neurosurgery 2013; 71:741-8; discussion 748. [PMID: 22653387 DOI: 10.1227/neu.0b013e318260fd73] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Glioblastoma is a fatal brain tumor in needing urgent effective therapy. Treatments with both oncolytic viruses and immunotherapy have shown preclinical efficacy and clinical promise. We sought to exploit possible synergies between oncolytic herpes simplex virus type 1 (oHSV-1) infection of intracranial gliomas and delivery of immune-stimulating fms-like tyrosine kinase 3 ligand (Flt3L) by engineering a herpes vector to express the cytokine. OBJECTIVE To construct an oHSV-1 vector that expresses high levels of Flt3L and examine its antiglioma efficacy in an immunocompetent murine model. METHODS G47Δ and a bacterial artificial chromosome system were used to generate a novel oHSV-1, termed G47Δ-Flt3L, expressing Flt3L. Cytokine expression was confirmed, and G47Δ-Flt3L was injected intratumorally into established intracranial CT-2A gliomas in syngeneic C57/Bl6 mice. Animals were followed for survival and assessed by the Kaplan-Meier method. RESULTS G47Δ-Flt3L expressed high levels of Flt3L in culture. Expression of Flt3L affected neither viral replication nor had a cytotoxic effect on CT2A glioma cells. Direct inoculation into intracerebral CT2A glioma cells resulted in high levels of detectable Flt3L in mouse blood and was superior to parental G47Δ in prolonging survival in glioma-bearing animals. CONCLUSION Treatment with G47Δ-Flt3L improves survival of glioma-bearing mice.
Collapse
Affiliation(s)
- Zachary Barnard
- Simches Brain Tumor Research Laboratories, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, USA
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Taylor TE, Furnari FB, Cavenee WK. Targeting EGFR for treatment of glioblastoma: molecular basis to overcome resistance. Curr Cancer Drug Targets 2012; 12:197-209. [PMID: 22268382 DOI: 10.2174/156800912799277557] [Citation(s) in RCA: 172] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Revised: 05/30/2011] [Accepted: 06/27/2011] [Indexed: 12/17/2022]
Abstract
Glioblastoma (glioblastoma multiforme; GBM; WHO Grade IV) accounts for the majority of primary malignant brain tumors in adults. Amplification and mutation of the epidermal growth factor receptor (EGFR) gene represent signature genetic abnormalities encountered in GBM. A range of potential therapies that target EGFR or its mutant constitutively active form, ΔEGFR, including tyrosine kinase inhibitors (TKIs), monoclonal antibodies, vaccines, and RNA-based agents, are currently in development or in clinical trials for the treatment of GBM. Data from experimental studies evaluating these therapies have been very promising; however, their efficacy in the clinic has so far been limited by both upfront and acquired drug resistance. This review discusses the current status of anti-EGFR agents and the recurrent problem of resistance to these agents that strongly indicates that a multiple target approach will provide a more favorable future for these types of targeted therapies in GBM.
Collapse
Affiliation(s)
- T E Taylor
- Ludwig Institute for Cancer Research, La Jolla, CA, USA
| | | | | |
Collapse
|
16
|
Kushchayev SV, Sankar T, Eggink LL, Kushchayeva YS, Wiener PC, Hoober JK, Eschbacher J, Liu R, Shi FD, Abdelwahab MG, Scheck AC, Preul MC. Monocyte galactose/N-acetylgalactosamine-specific C-type lectin receptor stimulant immunotherapy of an experimental glioma. Part 1: stimulatory effects on blood monocytes and monocyte-derived cells of the brain. Cancer Manag Res 2012; 4:309-23. [PMID: 23049280 PMCID: PMC3459590 DOI: 10.2147/cmar.s33248] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Objectives Immunotherapy with immunostimulants is an attractive therapy against gliomas. C-type lectin receptors specific for galactose/N-acetylgalactosamine (GCLR) regulate cellular differentiation, recognition, and trafficking of monocyte-derived cells. A peptide mimetic of GCLR ligands (GCLRP) was used to activate blood monocytes and populations of myeloid-derived cells against a murine glioblastoma. Methods The ability of GCLRP to stimulate phagocytosis by human microglia and monocyte-derived cells of the brain (MDCB) isolated from a human glioblastoma was initially assessed in vitro. Induction of activation markers on blood monocytes was assayed by flow cytometry after administration of GCLRP to naive mice. C57BL/6 mice underwent stereotactic intracranial implantation of GL261 glioma cells and were randomized for tumor size by magnetic resonance imaging, which was also used to assess increase in tumor size. Brain tumor tissues were analyzed using flow cytometry, histology, and enzyme-linked immunosorbent assay with respect to tumor, peritumoral area, and contralateral hemisphere regions. Results GCLRP exhibited strong stimulatory effect on MDCBs and blood monocytes in vitro and in vivo. GCLRP was associated with an increased percentage of precursors of dendritic cells in the blood (P = 0.003), which differentiated into patrolling macrophages in tumoral (P = 0.001) and peritumoral areas (P = 0.04), rather than into dendritic cells, as in control animals. Treatment with GCLRP did not result in a significant change in survival of mice bearing a tumor. Conclusions In vitro and in vivo activation of monocytes was achieved by administration of GCLR to mice. GCLRP-activated blood monocytes were recruited to the brain and exhibited specific phenotypes corresponding with tumor region (glioma, peritumoral zone, and contralateral glioma-free hemisphere). GCLRP treatment alone was associated with increased glioma mass as the result of the infiltration of phagocytic cells. Regional specificity for MDCB may have significant tumor treatment implications.
Collapse
Affiliation(s)
- Sergiy V Kushchayev
- Neurosurgery Research Laboratory, Barrow Neurological Institute, St Joseph's Hospital and Medical Center, Phoenix
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Cellular-based immunotherapies for patients with glioblastoma multiforme. Clin Dev Immunol 2012; 2012:764213. [PMID: 22474481 PMCID: PMC3299309 DOI: 10.1155/2012/764213] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Accepted: 12/08/2011] [Indexed: 12/30/2022]
Abstract
Treatment of patients with glioblastoma multiforme (GBM) remains to be a challenge with a median survival of 14.6 months following diagnosis. Standard treatment options include surgery, radiation therapy, and systemic chemotherapy with temozolomide. Despite the fact that the brain constitutes an immunoprivileged site, recent observations after immunotherapies with lysate from autologous tumor cells pulsed on dendritic cells (DCs), peptides, protein, messenger RNA, and cytokines suggest an immunological and even clinical response from immunotherapies. Given this plethora of immunomodulatory therapies, this paper gives a structure overview of the state-of-the art in the field. Particular emphasis was also put on immunogenic antigens as potential targets for a more specific stimulation of the immune system against GBM.
Collapse
|
18
|
Abstract
Glioblastoma, the most aggressive primary brain tumor, thrives in a microenvironment of relative immunosuppression within the relatively immune-privileged central nervous system. Despite treatments with surgery, radiation therapy, and chemotherapy, prognosis remains poor. The recent success of immunotherapy in the treatment of other cancers has renewed interest in vaccine therapy for the treatment of gliomas. In this article, we outline various immunotherapeutic strategies, review recent clinical trials data, and discuss the future of vaccine therapy for glioblastoma.
Collapse
Affiliation(s)
- Alissa A. Thomas
- Department of Neurology, Dartmouth Medical School and Dartmouth-Hitchcock Medical Center, Lebanon NH 03756
| | - Marc S. Ernstoff
- Department of Medicine, Section of Hematology/Oncology, Dartmouth Medical School and Dartmouth-Hitchcock Medical Center, Lebanon NH 03756
- Medical Oncology Immunotherapy Program, Dartmouth Medical School and Dartmouth-Hitchcock Medical Center, Lebanon NH 03756
| | - Camilo E. Fadul
- Department of Medicine, Section of Hematology/Oncology, Dartmouth Medical School and Dartmouth-Hitchcock Medical Center, Lebanon NH 03756
- Department of Neurology, Dartmouth Medical School and Dartmouth-Hitchcock Medical Center, Lebanon NH 03756
- Medical Oncology Immunotherapy Program, Dartmouth Medical School and Dartmouth-Hitchcock Medical Center, Lebanon NH 03756
- Neuro-oncology Program, Norris Cotton Cancer Center, Dartmouth Medical School and Dartmouth-Hitchcock Medical Center, Lebanon NH 03756
| |
Collapse
|
19
|
Wiwanitkit V. T-cell epitope finding on EPHA2 for further glioma vaccine development: An immunomics study. J Pediatr Neurosci 2011; 6:2-3. [PMID: 21977079 PMCID: PMC3173908 DOI: 10.4103/1817-1745.84398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background: Glioma is a deadly neurological tumor. For modern management of glioma, glioma vaccinotherapy is the new concept. Materials and Methods: Based on present biomedical technique, the identification of T-cell epitopes via MHC mapping can help clarify the inter-relationship of tumor and immune system. This process can be performed using advanced immunoinformatics technique. Results: Here, the author performs an immunoinformatics analysis to find alternative epitopes for glioma-related antigen, EPHA2. Conclusion: After complete manipulation on EPHA2 molecules, the five best epitopes were derived.
Collapse
|