51
|
Buchroithner J, Erhart F, Pichler J, Widhalm G, Preusser M, Stockhammer G, Nowosielski M, Iglseder S, Freyschlag CF, Oberndorfer S, Bordihn K, von Campe G, Hoffermann M, Ruckser R, Rössler K, Spiegl-Kreinecker S, Fischer MB, Czech T, Visus C, Krumpl G, Felzmann T, Marosi C. Audencel Immunotherapy Based on Dendritic Cells Has No Effect on Overall and Progression-Free Survival in Newly Diagnosed Glioblastoma: A Phase II Randomized Trial. Cancers (Basel) 2018; 10:E372. [PMID: 30301187 PMCID: PMC6210090 DOI: 10.3390/cancers10100372] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 09/22/2018] [Accepted: 09/30/2018] [Indexed: 11/17/2022] Open
Abstract
Dendritic cells (DCs) are antigen-presenting cells that are capable of priming anti-tumor immune responses, thus serving as attractive tools to generate tumor vaccines. In this multicentric randomized open-label phase II study, we investigated the efficacy of vaccination with tumor lysate-charged autologous DCs (Audencel) in newly diagnosed glioblastoma multiforme (GBM). Patients aged 18 to 70 years with histologically proven primary GBM and resection of at least 70% were randomized 1:1 to standard of care (SOC) or SOC plus vaccination (weekly intranodal application in weeks seven to 10, followed by monthly intervals). The primary endpoint was progression-free survival at 12 months. Secondary endpoints were overall survival, safety, and toxicity. Seventy-six adult patients were analyzed in this study. Vaccinations were given for seven (3⁻20) months on average. No severe toxicity was attributable to vaccination. Seven patients showed flu-like symptoms, and six patients developed local skin reactions. Progression-free survival at 12 months did not differ significantly between the control and vaccine groups (28.4% versus 24.5%, p = 0.9975). Median overall survival was similar with 18.3 months (vaccine: 564 days, 95% CI: 436⁻671 versus control: 568 days, 95% CI: 349⁻680; p = 0.89, harzard ratio (HR) 0.99). Hence, in this trial, the clinical outcomes of patients with primary GBM could not be improved by the addition of Audencel to SOC.
Collapse
Affiliation(s)
- Johanna Buchroithner
- University Clinic for Neurosurgery, Kepler University Hospital, Johannes Kepler University, Wagner-Jauregg-Weg 15, 4020 Linz, Austria.
| | - Friedrich Erhart
- Department of Neurosurgery, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.
- Institute of Neurology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| | - Josef Pichler
- Department of Internal Medicine and Neurooncology, Kepler University Hospital, Johannes Kepler University, Wagner-Jauregg-Weg 15, 4020 Linz, Austria.
| | - Georg Widhalm
- Department of Neurosurgery, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| | - Matthias Preusser
- Clinical Division of Medical Oncology, Department for Internal Medicine I, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| | - Günther Stockhammer
- Department of Neurology, Medical University of Innsbruck, Christoph-Probst-Platz 1, Innrain 52, 6020 Innsbruck, Austria.
| | - Martha Nowosielski
- Department of Neurology, Medical University of Innsbruck, Christoph-Probst-Platz 1, Innrain 52, 6020 Innsbruck, Austria.
| | - Sarah Iglseder
- Department of Neurology, Medical University of Innsbruck, Christoph-Probst-Platz 1, Innrain 52, 6020 Innsbruck, Austria.
| | - Christian F Freyschlag
- Department of Neurosurgery, Medical University of Innsbruck, Christoph-Probst-Platz 1, Innrain 52, 6020 Innsbruck, Austria.
| | - Stefan Oberndorfer
- Department of Neurology, University Clinic St. Pölten, Karl Landsteiner Privat Universität, Dunant-Platz 1, 3100 St. Pölten, Austria.
| | - Karin Bordihn
- Department of Neurosurgery, Landeskrankenhaus Salzburg, University Clinic of the Paracelsus Private Medical University, Müllner Hauptstraße 48, 5020 Salzburg, Austria.
| | - Gord von Campe
- Department of Neurosurgery, Medical University of Graz, Auenbruggerplatz 29, 8036 Graz, Austria.
| | - Markus Hoffermann
- Department of Neurosurgery, Medical University of Graz, Auenbruggerplatz 29, 8036 Graz, Austria.
| | - Reinhard Ruckser
- Department of Internal Medicine 2, Donauspital, Langobardenstraße 122, 1220 Vienna, Austria.
| | - Karl Rössler
- Department of Neurosurgery, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 6, 91054 Erlangen, Germany.
| | - Sabine Spiegl-Kreinecker
- University Clinic for Neurosurgery, Kepler University Hospital, Johannes Kepler University, Wagner-Jauregg-Weg 15, 4020 Linz, Austria.
| | - Michael B Fischer
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| | - Thomas Czech
- Department of Neurosurgery, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| | - Carmen Visus
- Activartis Biotech GmbH, Wilhelminenstraße 91/IIf, 1160 Vienna, Austria.
| | - Günther Krumpl
- Activartis Biotech GmbH, Wilhelminenstraße 91/IIf, 1160 Vienna, Austria.
| | - Thomas Felzmann
- Activartis Biotech GmbH, Wilhelminenstraße 91/IIf, 1160 Vienna, Austria.
| | - Christine Marosi
- Clinical Division of Medical Oncology, Department for Internal Medicine I, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria.
| |
Collapse
|
52
|
Comparative effect of immunotherapy and standard therapy in patients with high grade glioma: a meta-analysis of published clinical trials. Sci Rep 2018; 8:11800. [PMID: 30087385 PMCID: PMC6081409 DOI: 10.1038/s41598-018-30296-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 07/20/2018] [Indexed: 01/26/2023] Open
Abstract
Immunotherapy holds great promise in the treatment of high grade glioma (HGG). We performed a comprehensive meta-analysis of clinical trials involving dendritic cell (DC) therapy and viral therapy (VT) for the treatment of HGG, in order to assess their clinical impact in comparison to standard treatments in terms of overall survival (OS) and progression-free survival (PFS). To our knowledge, this is the first meta-analysis to evaluate VT for the treatment of HGG, allowing comparison of different immunotherapeutic approaches. Thirteen eligible studies of 1043 cases were included in the meta-analysis. For DC vaccination, in terms of OS, both newly diagnosed patients (HR, 0.65) and patients who suffered from recurrent HGGs (HR = 0.63) presented markedly improved results compared to the control groups. PFS was also improved (HR = 0.49) but was not statistically significant (p = 0.1). A slight improvement was observed for newly diagnosed patients receiving VT in terms of OS (HR = 0.88) while PFS was inferior for patients in the experimental arm (HR = 1.16). Our results show that DC therapy greatly improves OS for patients with both newly diagnosed and recurrent HGGs. VT, however, did not provide any statistically significant improvements in terms of OS and PFS for patients with newly diagnosed HGGs.
Collapse
|
53
|
High-grade glioma associated immunosuppression does not prevent immune responses induced by therapeutic vaccines in combination with T reg depletion. Cancer Immunol Immunother 2018; 67:1545-1558. [PMID: 30054667 PMCID: PMC6182405 DOI: 10.1007/s00262-018-2214-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 07/20/2018] [Indexed: 01/20/2023]
Abstract
High-grade gliomas (HGG) exert systemic immunosuppression, which is of particular importance as immunotherapeutic strategies such as therapeutic vaccines are increasingly used to treat HGGs. In a first cohort of 61 HGG patients we evaluated a panel of 30 hematological and 34 plasma biomarkers. Then, we investigated in a second cohort of 11 relapsed HGG patients receiving immunomodulation with metronomic cyclophosphamide upfront to a DC-based vaccine whether immune abnormalities persisted and whether they hampered induction of IFNγ+ T-cell responses. HGG patients from the first cohort showed increased numbers of leukocytes, neutrophils and MDSCs and in parallel reduced numbers of CD4+/CD8+ T-cells, plasmacytoid and conventional DC2s. MDSCs and T-cell alterations were more profound in WHO IV° glioma patients. Moreover, levels of MDSCs and epidermal growth factor were negatively associated with survival. Serum levels of IL-2, IL-4, IL-5 and IL-10 were altered in HGG patients, however, without any impact on clinical outcome. In the immunotherapy cohort, 6-month overall survival was 100%. Metronomic cyclophosphamide led to > 40% reduction of regulatory T cells (Treg). In parallel to Treg-depletion, MDSCs and DC subsets became indistinguishable from healthy controls, whereas T-lymphopenia persisted. Despite low T-cells, IFNγ-responses could be induced in 9/10 analyzed cases. Importantly, frequency of CD8+VLA-4+ T-cells with CNS-homing properties, but not of CD4+ VLA-4+ T-cells, increased during vaccination. Our study identifies several features of systemic immunosuppression in HGGs. Metronomic cyclophosphamide in combination with an active immunization alleviates the latter and the combined treatment allows induction of a high rate of anti-glioma immune responses.
Collapse
|
54
|
Eagles ME, Nassiri F, Badhiwala JH, Suppiah S, Almenawer SA, Zadeh G, Aldape KD. Dendritic cell vaccines for high-grade gliomas. Ther Clin Risk Manag 2018; 14:1299-1313. [PMID: 30100728 PMCID: PMC6067774 DOI: 10.2147/tcrm.s135865] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma (GBM) is the most common and fatal primary adult brain tumor. To date, various promising chemotherapeutic regimens have been trialed for use in GBM; however, temozolomide (TMZ) therapy remains the only US Food and Drug Administration-approved first-line chemotherapeutic option for newly diagnosed GBM. Despite maximal therapy with surgery and combined concurrent chemoradiation and adjuvant TMZ therapy, the median overall survival remains approximately 14 months. Given the failure of conventional chemotherapeutic strategies in GBM, there has been renewed interest in the role of immunotherapy in GBM. Dendritic cells are immune antigen-presenting cells that play a role in both the innate and adaptive immune system, thereby making them prime vehicles for immunotherapy via dendritic cell vaccinations (DCVs) in various cancers. There is great enthusiasm surrounding the use of DCVs for GBM with multiple ongoing trials. In this review, we comprehensively summarize the safety, efficacy, and quality of life results from 33 trials reporting on DCV for high-grade gliomas.
Collapse
Affiliation(s)
- Matthew E Eagles
- Section of Neurosurgery, Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Farshad Nassiri
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada, .,MacFeeters-Hamilton Neuro-Oncology Program, University Health Network, Toronto, ON, Canada
| | - Jetan H Badhiwala
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada,
| | - Suganth Suppiah
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada,
| | - Saleh A Almenawer
- Division of Neurosurgery, Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Gelareh Zadeh
- MacFeeters-Hamilton Neuro-Oncology Program, University Health Network, Toronto, ON, Canada.,Division of Neurosurgery, University Health Network, Toronto, ON, Canada
| | - Kenneth D Aldape
- MacFeeters-Hamilton Neuro-Oncology Program, University Health Network, Toronto, ON, Canada.,Division of Pathology, University Health Network, Toronto, ON, Canada
| |
Collapse
|
55
|
Rapp M, Grauer OM, Kamp M, Sevens N, Zotz N, Sabel M, Sorg RV. A randomized controlled phase II trial of vaccination with lysate-loaded, mature dendritic cells integrated into standard radiochemotherapy of newly diagnosed glioblastoma (GlioVax): study protocol for a randomized controlled trial. Trials 2018; 19:293. [PMID: 29801515 PMCID: PMC5970474 DOI: 10.1186/s13063-018-2659-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 05/02/2018] [Indexed: 01/06/2023] Open
Abstract
Background Despite the combination of surgical resection, radio- and chemotherapy, median survival of glioblastoma multiforme (GBM) patients only slightly increased in the last years. Disease recurrence is definite with no effective therapy existing after tumor removal. Dendritic cell (DC) vaccination is a promising active immunotherapeutic approach. There is clear evidence that it is feasible, results in immunological anti-tumoral responses, and appears to be beneficial for survival and quality of life of GBM patients. Moreover, combining it with the standard therapy of GBM may allow exploiting synergies between the treatment modalities. In this randomized controlled trial, we seek to confirm these promising initial results. Methods One hundred and thirty-six newly diagnosed, isocitrate dehydrogenase wildtype GBM patients will be randomly allocated (1:1 ratio, stratified by O6-methylguanine-DNA-methyltransferase promotor methylation status) after near-complete resection in a multicenter, prospective phase II trial into two groups: (1) patients receiving the current therapeutic “gold standard” of radio/temozolomide chemotherapy and (2) patients receiving DC vaccination as an add-on to the standard therapy. A recruitment period of 30 months is anticipated; follow-up will be 2 years. The primary objective of the study is to compare overall survival (OS) between the two groups. Secondary objectives are comparing progression-free survival (PFS) and 6-, 12- and 24-month OS and PFS rates, the safety profile, overall and neurological performance and quality of life. Discussion Until now, close to 500 GBM patients have been treated with DC vaccination in clinical trials or on a compassionate-use basis. Results have been encouraging, but cannot provide robust evidence of clinical efficacy because studies have been non-controlled or patient numbers have been low. Therefore, a prospective, randomized phase II trial with a sufficiently large number of patients is now mandatory for clear evidence regarding the impact of DC vaccination on PFS and OS in GBM. Trial registration Protocol code: GlioVax, date of registration: 17. February 2017. Trial identifier: EudraCT-Number 2017–000304-14. German Registry for Clinical Studies, ID: DRKS00013248 (approved primary register in the WHO network) and at ClinicalTrials.gov, ID: NCT03395587. Registered on 11 March 2017. Electronic supplementary material The online version of this article (10.1186/s13063-018-2659-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marion Rapp
- Department of Neurosurgery, Heinrich Heine University Hospital, Moorenstr. 5, 40225, Düsseldorf, Germany. .,Department of Neurosurgery, Heinrich Heine University Hospital Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.
| | - Oliver M Grauer
- Department of Neurology, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Marcel Kamp
- Department of Neurosurgery, Heinrich Heine University Hospital, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Natalie Sevens
- Department of Neurosurgery, Heinrich Heine University Hospital, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Nikola Zotz
- Coordination Center for Clinical Trials, Heinrich Heine University Hospital, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Michael Sabel
- Department of Neurosurgery, Heinrich Heine University Hospital, Moorenstr. 5, 40225, Düsseldorf, Germany
| | - Rüdiger V Sorg
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich Heine University Hospital, Moorenstr. 5, 40225, Düsseldorf, Germany
| |
Collapse
|
56
|
Ameratunga M, Coleman N, Welsh L, Saran F, Lopez J. CNS cancer immunity cycle and strategies to target this for glioblastoma. Oncotarget 2018; 9:22802-22816. [PMID: 29854316 PMCID: PMC5978266 DOI: 10.18632/oncotarget.24896] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 03/11/2018] [Indexed: 02/06/2023] Open
Abstract
Immunotherapeutics have revolutionized the management of solid malignancies over the last few years. Nevertheless, despite relative successes of checkpoint inhibitors in numerous solid tumour types, success in tumours of the central nervous system (CNS) has been lacking. There are several possible reasons for the relative lack of success of immunotherapeutics in this setting, including the immune microenvironment of glioblastoma, lymphocyte tracking through the blood-brain barrier (BBB) into the central nervous system and impairment of drug delivery into the CNS through the BBB. This review utilizes the cancer-immunity cycle as a conceptual framework through which the specific challenges associated with the development of immunotherapeutics for CNS malignancies can be viewed.
Collapse
Affiliation(s)
- Malaka Ameratunga
- Drug Development Unit, Royal Marsden Hospital and The Institute of Cancer Research, Sutton SM2 5PT, UK
| | - Niamh Coleman
- Drug Development Unit, Royal Marsden Hospital and The Institute of Cancer Research, Sutton SM2 5PT, UK
| | - Liam Welsh
- Department of Neuro-Oncology, Royal Marsden Hospital and The Institute of Cancer Research, Sutton SM2 5PT, UK
| | - Frank Saran
- Department of Neuro-Oncology, Royal Marsden Hospital and The Institute of Cancer Research, Sutton SM2 5PT, UK
| | - Juanita Lopez
- Drug Development Unit, Royal Marsden Hospital and The Institute of Cancer Research, Sutton SM2 5PT, UK
| |
Collapse
|
57
|
Tran TH, Tran TTP, Nguyen HT, Phung CD, Jeong JH, Stenzel MH, Jin SG, Yong CS, Truong DH, Kim JO. Nanoparticles for dendritic cell-based immunotherapy. Int J Pharm 2018; 542:253-265. [PMID: 29555438 DOI: 10.1016/j.ijpharm.2018.03.029] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 03/13/2018] [Accepted: 03/15/2018] [Indexed: 12/19/2022]
Abstract
Crosstalk among immune cells has attracted considerable attention with the advent of immunotherapy as a novel therapeutic approach for challenging diseases, especially cancer, which is the leading cause of mortality worldwide. Dendritic cells-the key antigen-presenting cells-play a pivotal role in immunological response by presenting exogenous epitopes to T cells, which induces the self-defense mechanisms of the body. Furthermore, nanotechnology has provided promising ways for diagnosing and treating cancer in the last decade. The progress in nanoparticle drug carrier development, combined with enhanced understanding of the immune system, has enabled harnessing of anti-tumor immunity. This review focuses on the recent advances in nanotechnology that have improved the therapeutic efficacy of immunotherapies, with emphasis on dendritic cell physiology and its role in presenting antigens and eliciting therapeutic T cell response.
Collapse
Affiliation(s)
- Tuan Hiep Tran
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City, Vietnam; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam.
| | - Thi Thu Phuong Tran
- The Institute of Molecular Genetics of Montpellier, CNRS, Montpellier, France
| | - Hanh Thuy Nguyen
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Republic of Korea
| | - Cao Dai Phung
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Republic of Korea
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Republic of Korea
| | - Martina H Stenzel
- Centre for Advanced Macromolecular Design, School of Chemistry, University of New South Wales, Kensington, NSW 2052, Australia
| | - Sung Giu Jin
- Department of Pharmaceutical Engineering, Dankook University, 119 Dandae-ro, Cheonan 31116, Republic of Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Republic of Korea
| | - Duy Hieu Truong
- Institute of Research and Development, Duy Tan University, 03 Quang Trung, Da Nang, Vietnam.
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, 280 Daehak-ro, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|
58
|
Nazarkina ZK, Zajakina A, Laktionov PP. Maturation and Antigen Loading Protocols Influence Activity of Anticancer Dendritic Cells. Mol Biol 2018. [DOI: 10.1134/s0026893317050132] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
59
|
Madsen SJ, Christie C, Huynh K, Peng Q, Uzal FA, Krasieva TB, Hirschberg H. Limiting glioma development by photodynamic therapy-generated macrophage vaccine and allo-stimulation: an in vivo histological study in rats. JOURNAL OF BIOMEDICAL OPTICS 2018; 23:1-7. [PMID: 29417766 PMCID: PMC5802332 DOI: 10.1117/1.jbo.23.2.028001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 01/19/2018] [Indexed: 06/08/2023]
Abstract
Immunotherapy of brain tumors involves the stimulation of an antitumor immune response. This type of therapy can be targeted specifically to tumor cells thus sparing surrounding normal brain. Due to the presence of the blood-brain barrier, the brain is relatively isolated from the systemic circulation and, as such, the initiation of significant immune responses is more limited than other types of cancers. The purpose of this study was to show that the efficacy of tumor primed antigen presenting macrophage (MaF98) vaccines can be increased by: (1) photodynamic therapy (PDT) of the priming tumor cells and (2) intracranial injection of allogeneic glioma cells directly into the tumor site. Experiments were conducted in an in vivo brain tumor development model using Fischer rats and F98 (syngeneic) and BT4C (allogeneic) glioma cells. The results showed that immunization with Ma (acting as antigen-presenting cells), primed with PDT-treated tumor cells (MaF98), significantly slowed but did not prevent the growth of F98-induced tumors in the brain. Complete suppression of tumor development was obtained via MaF98 inoculation combined with direct intracranial injection of allogeneic glioma cells. No deleterious effects were noted in any of the animals during the 14-day observation period.
Collapse
Affiliation(s)
- Steen J. Madsen
- University of Nevada, Department of Health Physics and Diagnostic Sciences, Las Vegas, Nevada, United States
| | - Catherine Christie
- University of California, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
| | - Khoi Huynh
- University of California, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
| | - Qian Peng
- University of Oslo, Pathology Clinic, Rikshospitalet-Radiumhospitalet HF Medical Center, Montebello, Oslo, Norway
| | - Francisco A. Uzal
- University of California, School of Veterinary Medicine, Davis, San Bernardino, California, United States
| | - Tatiana B. Krasieva
- University of California, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
| | - Henry Hirschberg
- University of California, Beckman Laser Institute and Medical Clinic, Irvine, California, United States
| |
Collapse
|
60
|
Maxwell R, Luksik AS, Garzon-Muvdi T, Lim M. The Potential of Cellular- and Viral-Based Immunotherapies for Malignant Glioma-Dendritic Cell Vaccines, Adoptive Cell Transfer, and Oncolytic Viruses. Curr Neurol Neurosci Rep 2017; 17:50. [PMID: 28488122 DOI: 10.1007/s11910-017-0754-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW Malignant gliomas, including glioblastoma and anaplastic astrocytoma, are the most frequent primary brain tumors and present with many treatment challenges. In this review, we discuss the potential of cellular- and viral-based immunotherapies in the treatment of malignant glioma, specifically focusing on dendritic cell vaccines, adoptive cell therapy, and oncolytic viruses. RECENT FINDINGS Diverse cellular- and viral-based strategies have been engineered and optimized to generate either a specific or broad antitumor immune response in malignant glioma. Due to their successes in the preclinical arena, many of these therapies have undergone phase I and II clinical testing. These early clinical trials have demonstrated the feasibility, safety, and efficacy of these immunotherapies. Dendritic cell vaccines, adoptive cell transfer, and oncolytic viruses may have a potential role in the treatment of malignant glioma. However, these modalities must be investigated in well-designed phase III trials to prove their efficacy.
Collapse
Affiliation(s)
- Russell Maxwell
- Department of Neurosurgery, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Neurosurgery - Phipps 123, Baltimore, MD, 21287, USA
| | - Andrew S Luksik
- Department of Neurosurgery, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Neurosurgery - Phipps 123, Baltimore, MD, 21287, USA
| | - Tomas Garzon-Muvdi
- Department of Neurosurgery, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Neurosurgery - Phipps 123, Baltimore, MD, 21287, USA
| | - Michael Lim
- Department of Neurosurgery, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Neurosurgery - Phipps 123, Baltimore, MD, 21287, USA.
| |
Collapse
|
61
|
Panek WK, Kane JR, Young JS, Rashidi A, Kim JW, Kanojia D, Lesniak MS. Hitting the nail on the head: combining oncolytic adenovirus-mediated virotherapy and immunomodulation for the treatment of glioma. Oncotarget 2017; 8:89391-89405. [PMID: 29179527 PMCID: PMC5687697 DOI: 10.18632/oncotarget.20810] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 08/26/2017] [Indexed: 12/31/2022] Open
Abstract
Glioblastoma is a highly aggressive malignant brain tumor with a poor prognosis and the median survival 14.6 months. Immunomodulatory proteins and oncolytic viruses represent two treatment approaches that have recently been developed for patients with glioblastoma that could extend patient survival and result in better treatment outcomes for patients with this disease. Together, these approaches could potentially augment the treatment efficacy and strength of these anti-tumor therapies. In addition to oncolytic activities, this combinatory approach introduces immunomodulation locally only where cancerous cells are present. This thereby results in the change of the tumor microenvironment from immune-suppressive to immune-vulnerable via activation of cytotoxic T cells or through the removal of glioma cells immune-suppressive capability. This review discusses the strengths and weaknesses of adenoviral oncolytic therapy, and highlights the genetic modifications that result in more effective and targeted viral agents. Additionally, the mechanism of action of immune-activating agents is described and the results of previous clinical trials utilizing these treatments in other solid tumors are reviewed. The feasibility, synergy, and limitations for treatments that combine these two approaches are outlined and areas for which more work is needed are considered.
Collapse
Affiliation(s)
- Wojciech K Panek
- Department of Neurological Surgery, Northwestern University, Chicago, IL, 60611, USA
| | - J Robert Kane
- Department of Neurological Surgery, Northwestern University, Chicago, IL, 60611, USA
| | - Jacob S Young
- Pritzker School of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Aida Rashidi
- Department of Neurological Surgery, Northwestern University, Chicago, IL, 60611, USA
| | - Julius W Kim
- Department of Neurological Surgery, Northwestern University, Chicago, IL, 60611, USA
| | - Deepak Kanojia
- Department of Neurological Surgery, Northwestern University, Chicago, IL, 60611, USA
| | - Maciej S Lesniak
- Department of Neurological Surgery, Northwestern University, Chicago, IL, 60611, USA
| |
Collapse
|
62
|
Identification of T cell target antigens in glioblastoma stem-like cells using an integrated proteomics-based approach in patient specimens. Acta Neuropathol 2017; 134:297-316. [PMID: 28332095 DOI: 10.1007/s00401-017-1702-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 03/17/2017] [Accepted: 03/17/2017] [Indexed: 12/15/2022]
Abstract
Glioblastoma (GBM) is a highly aggressive brain tumor and still remains incurable. Among others, an immature subpopulation of self-renewing and therapy-resistant tumor cells-often referred to as glioblastoma stem-like cells (GSCs)-has been shown to contribute to disease recurrence. To target these cells personalized immunotherapy has gained a lot of interest, e.g. by reactivating pre-existing anti-tumor immune responses against GSC antigens. To identify T cell targets commonly presented by GSCs and their differentiated counterpart, we used a proteomics-based separation of GSC proteins in combination with a T cell activation assay. Altogether, 713 proteins were identified by LC-ESI-MS/MS mass spectrometry. After a thorough filtering process, 32 proteins were chosen for further analyses. Immunogenicity of corresponding peptides was tested ex vivo. A considerable number of these antigens induced T cell responses in GBM patients but not in healthy donors. Moreover, most of them were overexpressed in primary GBM and also highly expressed in recurrent GBM tissues. Interestingly, expression of the most frequent T cell target antigens could also be confirmed in quiescent, slow-cycling GSCs isolated in high purity by the DEPArray technology. Finally, for a subset of these T cell target antigens, an association between expression levels and higher T cell infiltration as well as an increased expression of positive immune modulators was observed. In summary, we identified novel immunogenic proteins, which frequently induce tumor-specific T cell responses in GBM patients and were also detected in vitro in therapy-resistant quiescent, slow-cycling GSCs. Stable expression of these T cell targets in primary and recurrent GBM support their suitability for future clinical use.
Collapse
|
63
|
Ogasawara M, Miyashita M, Iida M, Fukuhara T. Immunological Analysis and Generation of Dendritic Cells From Lavage Fluid Obtained by Cell-Free and Concentrated Ascites Reinfusion Therapy. Ther Apher Dial 2017; 21:255-262. [PMID: 28661089 DOI: 10.1111/1744-9987.12559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Cell-free and concentrated ascites reinfusion therapy (CART) is an effective treatment for patients with refractory ascites. Cellular components such as cancer cells and blood cells are removed and discarded. The aim of this study was to investigate the alteration of immune cells in lavage fluid and the generation of dendritic cells (DCs) from lavage fluid obtained by CART. Flow cytometry analysis showed a trend toward immunosuppression and impairment in innate immunity in lavage fluid. Immature DCs with downregulation of CD14 and increased antigen-uptake were generated by culturing monocytes obtained from lavage fluid with GM-CSF and IL4. Following the culture with proinflammatory mediators, mature DCs with upregulation of CD83 and potent ability of T cell activation were induced. There were no significant phenotypical or functional differences between these DCs and DCs derived from peripheral blood, indicating lavage fluid might be employed for an alternative cellular source for the generation of DCs.
Collapse
Affiliation(s)
| | - Mamiko Miyashita
- Institute for Artificial Organ, Transplantation and Gene Therapy, Sapporo Hokuyu Hospital, Sapporo, Japan
| | - Michio Iida
- Department of Palliative Medicine, Sapporo Kousei Hospital, Sapporo, Japan
| | - Takashi Fukuhara
- Department of Palliative Medicine, Sapporo Kousei Hospital, Sapporo, Japan
| |
Collapse
|
64
|
Wu X, Xu F, Liu J, Wang G. Comparative study of dendritic cells matured by using IL-1β, IL-6, TNF-α and prostaglandins E2 for different time span. Exp Ther Med 2017; 14:1389-1394. [PMID: 28810601 PMCID: PMC5526128 DOI: 10.3892/etm.2017.4649] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 04/11/2017] [Indexed: 12/03/2022] Open
Abstract
Interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α and prostaglandins E2 is considered as the standard cocktail for maturing dendritic cells (DCs). However, the appropriate time span for DC maturation with the standard cocktail remains unclear. The present study aimed to compare the differences between DCs matured with the standard cocktail for 24 and 48 h, respectively, and determine whether 24-h stimulation was sufficient for DC maturation. The findings demonstrated that, compared with DCs matured for 48 h, the levels of cluster of differentiation (CD)80, CD83, CD86 and programmed death-ligand 1 expression in DCs matured for 24 h were relatively lower. However, with the exception of CD80 whose mean fluorescence intensity (MFI) was higher in DCs matured for 48 h, the MFI values of other surface markers were comparable. Notably, the MFI of CD40 was higher in DCs matured for 24 h. In addition, the viability, T cell stimulatory capacity in allogeneic mixed lymphocyte reaction and cytokine production, including IL-12p40, IL-12p70 and IL-10, were all comparable between DCs matured for 24 and 48 h, respectively. These results indicated that 24-h stimulation may be sufficient for DC maturation when using the standard cocktail.
Collapse
Affiliation(s)
- Xuejie Wu
- Department of Infectious Diseases, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Feng Xu
- Department of Infectious Diseases, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Jinliang Liu
- Department of Infectious Diseases, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Guiqiang Wang
- Department of Infectious Diseases, Peking University First Hospital, Beijing 100034, P.R. China
| |
Collapse
|
65
|
Tao Z, Li S, Ichim TE, Yang J, Riordan N, Yenugonda V, Babic I, Kesari S. Cellular immunotherapy of cancer: an overview and future directions. Immunotherapy 2017; 9:589-606. [DOI: 10.2217/imt-2016-0086] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The clinical success of checkpoint inhibitors has led to a renaissance of interest in cancer immunotherapies. In particular, the possibility of ex vivo expanding autologous lymphocytes that specifically recognize tumor cells has attracted much research and clinical trial interest. In this review, we discuss the historical background of tumor immunotherapy using cell-based approaches, and provide some rationale for overcoming current barriers to success of autologous immunotherapy. An overview of adoptive transfer of lymphocytes, tumor infiltrating lymphocytes and dendritic cell therapies is provided. We conclude with discussing the possibility of gene-manipulating immune cells in order to augment therapeutic activity, including silencing of the immune-suppressive zinc finger orphan nuclear receptor, NR2F6, as an attractive means of overcoming tumor-associated immune suppression.
Collapse
Affiliation(s)
- Ziqi Tao
- The Affiliated XuZhou Center Hospital of Nanjing University of Chinese Medicine, The Affiliated XuZhou Hospital of Medical College of Southeast University, Jiangsu, China
| | - Shuang Li
- Department of Endocrinology, the Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | | | - Junbao Yang
- Department of Translational Neurosciences and Neurotherapeutics, Pacific Neuroscience Institute, John Wayne Cancer Institute, Providence Saint John’s Health Center, Santa Monica, CA 90404, USA
| | - Neil Riordan
- Medistem Panama, Inc., City of Knowledge, Clayton, Republic of Panama
| | - Venkata Yenugonda
- Department of Translational Neurosciences and Neurotherapeutics, Pacific Neuroscience Institute, John Wayne Cancer Institute, Providence Saint John’s Health Center, Santa Monica, CA 90404, USA
| | - Ivan Babic
- Department of Translational Neurosciences and Neurotherapeutics, Pacific Neuroscience Institute, John Wayne Cancer Institute, Providence Saint John’s Health Center, Santa Monica, CA 90404, USA
| | - Santosh Kesari
- Department of Translational Neurosciences and Neurotherapeutics, Pacific Neuroscience Institute, John Wayne Cancer Institute, Providence Saint John’s Health Center, Santa Monica, CA 90404, USA
- John Wayne Cancer Institute, 2200 Santa Monica Blvd, Santa Monica, CA 90404, USA
| |
Collapse
|
66
|
Inogés S, Tejada S, de Cerio ALD, Gállego Pérez-Larraya J, Espinós J, Idoate MA, Domínguez 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. J Transl Med 2017; 15:104. [PMID: 28499389 PMCID: PMC5427614 DOI: 10.1186/s12967-017-1202-z] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 05/03/2017] [Indexed: 01/20/2023] Open
Abstract
Background Prognosis of patients with glioblastoma multiforme (GBM) remains dismal, with median overall survival (OS) of about 15 months. It is therefore crucial to search alternative strategies that improve these results obtained with conventional treatments. In this context, immunotherapy seems to be a promising therapeutic option. We hypothesized that the addition of tumor lysate-pulsed autologous dendritic cells (DCs) vaccination to maximal safe resection followed by radiotherapy and concomitant and adjuvant temozolomide could improve patients’ survival. Methods We conducted a phase-II clinical trial of autologous DCs vaccination in patients with newly diagnosed patients GBM who were candidates to complete or near complete resection. Candidates were finally included if residual tumor volume was lower than 1 cc on postoperative radiological examination. Autologous DCs were generated from peripheral blood monocytes and pulsed with autologous whole tumor lysate. The vaccination calendar started before radiotherapy and was continued during adjuvant chemotherapy. Progression free survival (PFS) and OS were analyzed with the Kaplan–Meier method. Immune response were assessed in blood samples obtained before each vaccines. Results Thirty-two consecutive patients were screened, one of which was a screening failure due to insufficient resection. Median age was 61 years (range 42–70). Karnofsky performance score (KPS) was 90–100 in 29%, 80 in 35.5% and 60–70 in 35.5% of cases. MGMT (O6-methylguanine-DNA-methyltransferase) promoter was methylated in 45.2% of patients. No severe adverse effects related to immunotherapy were registered. Median PFS was 12.7 months (CI 95% 7–16) and median OS was 23.4 months (95% CI 16–33.1). Increase in post-vaccination tumor specific immune response after vaccines (proliferation or cytokine production) was detected in 11/27 evaluated patients. No correlation between immune response and survival was found. Conclusions Our results suggest that the addition of tumor lysate-pulsed autologous DCs vaccination to tumor resection and combined radio-chemotherapy is feasible and safe. A multicenter randomized clinical trial is warranted to evaluate the potential survival benefit of this therapeutic approach. Trial registration This phase-II trial was registered as EudraCT: 2009-009879-35 and ClinicalTrials.gov Identifier: NCT01006044 retrospectively registered Electronic supplementary material The online version of this article (doi:10.1186/s12967-017-1202-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Susana Inogés
- Cell Therapy Area, Clínica Universidad de Navarra, Avenida Pio XII 36, 31008, Pamplona, Navarra, Spain. .,Immunology and Immunotherapy Department, Clínica Universidad de Navarra, Avenida Pio XII 36, 31008, Pamplona, Navarra, Spain.
| | - Sonia Tejada
- Neurosurgery Department, Clínica Universidad de Navarra, Avenida Pio XII 36, 31008, Pamplona, Navarra, Spain
| | - Ascensión López-Díaz de Cerio
- Cell Therapy Area, Clínica Universidad de Navarra, Avenida Pio XII 36, 31008, Pamplona, Navarra, Spain.,Immunology and Immunotherapy Department, Clínica Universidad de Navarra, Avenida Pio XII 36, 31008, Pamplona, Navarra, Spain
| | | | - Jaime Espinós
- Oncology Department, Clínica Universidad de Navarra, Avenida Pio XII 36, 31008, Pamplona, Navarra, Spain
| | - Miguel Angel Idoate
- Pathology Department, Clínica Universidad de Navarra, Avenida Pio XII 36, 31008, Pamplona, Navarra, Spain
| | - Pablo Daniel Domínguez
- Radiology Department, Clínica Universidad de Navarra, Avenida Pio XII 36, 31008, Pamplona, Navarra, Spain
| | - Reyes García de Eulate
- Radiology Department, Clínica Universidad de Navarra, Avenida Pio XII 36, 31008, Pamplona, Navarra, Spain
| | - Javier Aristu
- Radiation Oncology Department, Clínica Universidad de Navarra, Avenida Pio XII 36, 31008, Pamplona, Navarra, Spain
| | - Maurizio Bendandi
- Section on Hematology/Oncology, Department of Internal Medicine, Comprehensive Cancer Center, Wake Forest University Baptist Healthcare Center, Winston-Salem, NC, USA.,Section of Hematology/Oncology, Department of Internal Medicine, W.G Hefner VA Medical Center, Salisbury/Charlotte, NC, USA
| | - Fernando Pastor
- Program of Molecular Therapies, Aptamer Unit, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Avenida Pio XII 55, 31008, Pamplona, Navarra, Spain
| | - Marta Alonso
- Program in Solid Tumors and Biomarkers, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Avenida Pio XII 55, 31008, Pamplona, Navarra, Spain
| | - Enrique Andreu
- Immunology and Immunotherapy Department, Clínica Universidad de Navarra, Avenida Pio XII 36, 31008, Pamplona, Navarra, Spain
| | - Felipe Prósper Cardoso
- Cell Therapy Area, Clínica Universidad de Navarra, Avenida Pio XII 36, 31008, Pamplona, Navarra, Spain.,Haematology and Haemotherapy Department, Clínica Universidad de Navarra, Avenida Pio XII 36, 31008, Pamplona, Navarra, Spain
| | - Ricardo Díez Valle
- Neurosurgery Department, Clínica Universidad de Navarra, Avenida Pio XII 36, 31008, Pamplona, Navarra, Spain.
| |
Collapse
|
67
|
Filley AC, Dey M. Dendritic cell based vaccination strategy: an evolving paradigm. J Neurooncol 2017; 133:223-235. [PMID: 28434112 DOI: 10.1007/s11060-017-2446-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 04/18/2017] [Indexed: 12/17/2022]
Abstract
Malignant gliomas (MG), tumors of glial origin, are the most commonly diagnosed primary intracranial malignancies in adults. Currently available treatments have provided only modest improvements in overall survival and remain limited by inevitable local recurrence, necessitating exploration of novel therapies. Among approaches being investigated, one of the leading contenders is immunotherapy, which aims to modulate immune pathways to stimulate the selective destruction of malignant cells. Dendritic cells (DCs) are potent initiators of adaptive immune responses and therefore crucial players in the development and success of immunotherapy. Clinical trials of various DC-based vaccinations have demonstrated the induction of anti-tumor immune responses and prolonged survival in the setting of many cancers. In this review, we summarize current literature regarding DCs and their role in the tumor microenvironment, their application and current clinical use in immunotherapy, current challenges limiting their efficacy in anti-cancer therapy, and future avenues for developing successful anti-tumor DC-based vaccines.
Collapse
Affiliation(s)
- Anna C Filley
- Department of Neurosurgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mahua Dey
- Department of Neurosurgery, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana University Purdue University Indianapolis (IUPUI), 320 W 15th Street, Neuroscience Building NB400A, Indianapolis, IN, 46202, USA.
| |
Collapse
|
68
|
Khoury HJ, Collins RH, Blum W, Stiff PS, Elias L, Lebkowski JS, Reddy A, Nishimoto KP, Sen D, Wirth ED, Case CC, DiPersio JF. Immune responses and long-term disease recurrence status after telomerase-based dendritic cell immunotherapy in patients with acute myeloid leukemia. Cancer 2017; 123:3061-3072. [PMID: 28411378 DOI: 10.1002/cncr.30696] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 02/21/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Telomerase activity in leukemic blasts frequently is increased among patients with high-risk acute myeloid leukemia (AML). In the current study, the authors evaluated the feasibility, safety, immunogenicity, and therapeutic potential of human telomerase reverse transcriptase (hTERT)-expressing autologous dendritic cells (hTERT-DCs) in adult patients with AML. METHODS hTERT-DCs were produced from patient-specific leukapheresis, electroporated with an mRNA-encoding hTERT and a lysosomal-targeting sequence, and cryopreserved. A total of 22 patients with a median age of 58 years (range, 30-75 years) with intermediate-risk or high-risk AML in first or second complete remission (CR) were enrolled. hTERT-DCs were generated for 24 patients (73%). A median of 17 intradermal vaccinations (range, 6-32 intradermal vaccinations) containing 1×107 cells were administered as 6 weekly injections followed by 6 biweekly injections. A total of 21 patients (16 in first CR, 3 in second CR, and 2 with early disease recurrence) received hTERT-DCs. RESULTS hTERT-DCs were well tolerated with no severe toxicities reported, with the exception of 1 patient who developed idiopathic thrombocytopenic purpura. Of the 19 patients receiving hTERT-DCs in CR, 11 patients (58%) developed hTERT-specific T-cell responses that primarily were targeted toward hTERT peptides with predicted low human leukocyte antigen (HLA)-binding affinities. With a median follow-up of 52 months, 58% of patients in CR (11 of 19 patients) were free of disease recurrence at the time of their last follow-up visit; 57% of the patients who were aged ≥60 years (4 of 7 patients) also were found to be free of disease recurrence at a median follow-up of 54 months. CONCLUSIONS The generation of hTERT-DCs is feasible and vaccination with hTERT-DCs appears to be safe and may be associated with favorable recurrence-free survival. Cancer 2017;123:3061-72. © 2017 American Cancer Society.
Collapse
Affiliation(s)
- Hanna J Khoury
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia
| | - Robert H Collins
- Department of Internal Medicine, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas
| | - William Blum
- Department of Hematology, The Ohio State University, Columbus, Ohio
| | - Patrick S Stiff
- Department of Medicine, Loyola University, Maywood, Illinois
| | | | | | | | | | - Debasish Sen
- Asterias Biotherapeutics Inc, Menlo Park, California
| | | | - Casey C Case
- Asterias Biotherapeutics Inc, Menlo Park, California
| | - John F DiPersio
- Department of Oncology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
69
|
Srinivasan VM, Ferguson SD, Lee S, Weathers SP, Kerrigan BCP, Heimberger AB. Tumor Vaccines for Malignant Gliomas. Neurotherapeutics 2017; 14:345-357. [PMID: 28389997 PMCID: PMC5398993 DOI: 10.1007/s13311-017-0522-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Despite continued research efforts, glioblastoma multiforme (GBM) remains the deadliest brain tumor. Immunotherapy offers a novel way to treat this disease, the genetic signature of which is not completely elucidated. Additionally, these tumors are known to induce immunosuppression in the surrounding tumor microenvironment via an array of mechanisms, making effective treatment all the more difficult. The immunotherapeutic strategy of using tumor vaccines offers a way to harness the activity of the host immune system to potentially control tumor progression. GBM vaccines can react to a variety of tumor-specific antigens, which can be harvested from the patient's unique pathological condition using selected immunotherapy techniques. This article reviews the rationale behind and development of GBM vaccines, the relevant clinical trials, and the challenges involved in this treatment strategy.
Collapse
Affiliation(s)
| | - Sherise D Ferguson
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Sungho Lee
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA
| | - Shiao-Pei Weathers
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
70
|
Sokratous G, Polyzoidis S, Ashkan K. Immune infiltration of tumor microenvironment following immunotherapy for glioblastoma multiforme. Hum Vaccin Immunother 2017; 13:2575-2582. [PMID: 28362548 DOI: 10.1080/21645515.2017.1303582] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Autologous dentritic cell immunotherapy has been proven effective in treating tumors outside the central nervous system. Current evidence from phase I and II trials suggest a similar efficacy for central nervous system tumors as well and that an active immune response against these tumors can be generated. We aim to review the literature to identify the types of immune responses against gliomas found to be generated by dendritic cell vaccinations and the types of immune cells subsequently infiltrating the glioma microenvironment. A systematic review of the literature was performed by searching the online databases PubMEd, Google Scholar, and EMBASE with use of the keywords intratumoral, infiltration, lymphocytic, vaccination and gliomas. Seven studies reporting lymphocytic infiltration of gliomas microenvironment were identified. Three studies (42.8%) reported presence of tumor infiltrating lymphocytes in 50%, 50% and 28.6% of included patients respectively in the post-vaccination specimens that were not present in the pre-vaccination samples. The remaining 4 (57.2%) reported an up to 6-fold increase in the number of pre-existing lymphocytes following vaccination. Present data indicate that tumor infiltration by lymphocytes can be induced by dentritic cell immunotherapy and that this may positively affect clinical outcome. It still remains unclear which factors influence the above reaction and therefore prediction of response to treatment is still not possible.
Collapse
Affiliation(s)
- Giannis Sokratous
- a Clinical Research Fellow, Department of Neurosurgery , King's College Hospital , Denmark Hill, London , UK
| | - Stavros Polyzoidis
- b Clinical Fellow, Department of Neurosurgery , King's College Hospital , Denmark Hill, London , UK.,c First Department of Neurosurgery - AHEPA Hospital , Aristotle University of Thessaloniki , Greece
| | - Keyoumars Ashkan
- d Professor of Neurosurgery, Department of Neurosurgery , King's College Hospital , Denmark Hill, London , UK
| |
Collapse
|
71
|
The development of dendritic cell vaccine-based immunotherapies for glioblastoma. Semin Immunopathol 2017; 39:225-239. [PMID: 28138787 DOI: 10.1007/s00281-016-0616-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 12/20/2016] [Indexed: 12/17/2022]
Abstract
In this review, we focus on the biologic advantages of dendritic cell-based vaccinations as a therapeutic strategy for cancer as well as preclinical and emerging clinical data associated with such approaches for glioblastoma patients.
Collapse
|
72
|
Farber SH, Elsamadicy AA, Atik AF, Suryadevara CM, Chongsathidkiet P, Fecci PE, Sampson JH. The Safety of available immunotherapy for the treatment of glioblastoma. Expert Opin Drug Saf 2017; 16:277-287. [PMID: 27989218 DOI: 10.1080/14740338.2017.1273898] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Glioblastoma (GBM) is the most common malignant primary brain tumor in adults. Current standard of care involves maximal surgical resection combined with adjuvant chemoradiation. Growing support exists for a role of immunotherapy in treating these tumors with the goal of targeted cytotoxicity. Here we review data on the safety for current immunotherapies being tested in GBM. Areas covered: Safety data from published clinical trials, including ongoing clinical trials were reviewed. Immunotherapeutic classes currently under investigation in GBM include various vaccination strategies, adoptive T cell immunotherapy, immune checkpoint blockade, monoclonal antibodies, and cytokine therapies. Trials include children, adolescents, and adults with either primary or recurrent GBM. Expert opinion: Based on the reviewed clinical trials, the current immunotherapies targeting GBM are safe and well-tolerated with minimal toxicities which should be noted. However, the gains in patient survival have been modest. A safe and well-tolerated combinatory immunotherapeutic approach may be essential for optimal efficacy towards GBM.
Collapse
Affiliation(s)
- S Harrison Farber
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b The Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , NC , USA
| | - Aladine A Elsamadicy
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b The Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , NC , USA
| | - Ahmet Fatih Atik
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b The Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , NC , USA
| | - Carter M Suryadevara
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b The Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , NC , USA.,c Department of Pathology , Duke University Medical Center , Durham , NC , USA
| | - Pakawat Chongsathidkiet
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b The Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , NC , USA.,c Department of Pathology , Duke University Medical Center , Durham , NC , USA
| | - Peter E Fecci
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b The Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , NC , USA.,c Department of Pathology , Duke University Medical Center , Durham , NC , USA
| | - John H Sampson
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b The Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , NC , USA.,c Department of Pathology , Duke University Medical Center , Durham , NC , USA
| |
Collapse
|
73
|
Harrison-Brown M, Liu GJ, Banati R. Checkpoints to the Brain: Directing Myeloid Cell Migration to the Central Nervous System. Int J Mol Sci 2016; 17:E2030. [PMID: 27918464 PMCID: PMC5187830 DOI: 10.3390/ijms17122030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/23/2016] [Accepted: 11/25/2016] [Indexed: 12/13/2022] Open
Abstract
Myeloid cells are a unique subset of leukocytes with a diverse array of functions within the central nervous system during health and disease. Advances in understanding of the unique properties of these cells have inspired interest in their use as delivery vehicles for therapeutic genes, proteins, and drugs, or as "assistants" in the clean-up of aggregated proteins and other molecules when existing drainage systems are no longer adequate. The trafficking of myeloid cells from the periphery to the central nervous system is subject to complex cellular and molecular controls with several 'checkpoints' from the blood to their destination in the brain parenchyma. As important components of the neurovascular unit, the functional state changes associated with lineage heterogeneity of myeloid cells are increasingly recognized as important for disease progression. In this review, we discuss some of the cellular elements associated with formation and function of the neurovascular unit, and present an update on the impact of myeloid cells on central nervous system (CNS) diseases in the laboratory and the clinic. We then discuss emerging strategies for harnessing the potential of site-directed myeloid cell homing to the CNS, and identify promising avenues for future research, with particular emphasis on the importance of untangling the functional heterogeneity within existing myeloid subsets.
Collapse
Affiliation(s)
- Meredith Harrison-Brown
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Health Sciences, The University of Sydney, Sydney, NSW 2141, Australia.
- Australian Nuclear Science and Technology Organisation, Sydney, NSW 2234, Australia.
| | - Guo-Jun Liu
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Health Sciences, The University of Sydney, Sydney, NSW 2141, Australia.
- Australian Nuclear Science and Technology Organisation, Sydney, NSW 2234, Australia.
| | - Richard Banati
- Discipline of Medical Imaging & Radiation Sciences, Faculty of Health Sciences, The University of Sydney, Sydney, NSW 2141, Australia.
- Australian Nuclear Science and Technology Organisation, Sydney, NSW 2234, Australia.
- Brain and Mind Centre, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
74
|
Artene SA, Turcu-Stiolica A, Hartley R, Ciurea ME, Daianu O, Brindusa C, Alexandru O, Tataranu LG, Purcaru SO, Dricu A. Dendritic cell immunotherapy versus bevacizumab plus irinotecan in recurrent malignant glioma patients: a survival gain analysis. Onco Targets Ther 2016; 9:6669-6677. [PMID: 27877052 PMCID: PMC5108618 DOI: 10.2147/ott.s112842] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background The bevacizumab and irinotecan protocol is considered a standard treatment regimen for recurrent malignant glioma. Recent advances in immunotherapy have hinted that vaccination with dendritic cells could become an alternative salvage therapy for the treatment of recurrent malignant glioma. Methods A search was performed on PubMed, Cochrane Library, Web of Science, ScienceDirect, and Embase in order to identify studies with patients receiving bevacizumab plus irinotecan or dendritic cell therapy for recurrent malignant gliomas. The data obtained from these studies were used to perform a systematic review and survival gain analysis. Results Fourteen clinical studies with patients receiving either bevacizumab plus irinotecan or dendritic cell vaccination were identified. Seven studies followed patients that received bevacizumab plus irinotecan (302 patients) and seven studies included patients that received dendritic cell immunotherapy (80 patients). For the patients who received bevacizumab plus irinotecan, the mean reported median overall survival was 7.5 (95% confidence interval [CI] 4.84–10.16) months. For the patients who received dendritic cell immunotherapy, the mean reported median overall survival was 17.9 (95% CI 11.34–24.46) months. For irinotecan + bevacizumab group, the mean survival gain was −0.02±2.00, while that for the dendritic cell immunotherapy group was −0.01±4.54. Conclusion For patients with recurrent malignant gliomas, dendritic cell immunotherapy treatment does not have a significantly different effect when compared with bevacizumab and irinotecan in terms of survival gain (P=0.535) and does not improve weighted survival gain (P=0.620).
Collapse
Affiliation(s)
| | | | | | | | | | | | - Oana Alexandru
- Department of Neurology, University of Medicine and Pharmacy of Craiova, Craiova
| | | | | | | |
Collapse
|
75
|
Antigen-specific immunoreactivity and clinical outcome following vaccination with glioma-associated antigen peptides in children with recurrent high-grade gliomas: results of a pilot study. J Neurooncol 2016; 130:517-527. [PMID: 27624914 DOI: 10.1007/s11060-016-2245-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 08/21/2016] [Indexed: 12/29/2022]
Abstract
Recurrent high-grade gliomas (HGGs) of childhood have an exceedingly poor prognosis with current therapies. Accordingly, new treatment approaches are needed. We initiated a pilot trial of vaccinations with peptide epitopes derived from glioma-associated antigens (GAAs) overexpressed in these tumors in HLA-A2+ children with recurrent HGG that had progressed after prior treatments. Peptide epitopes for three GAAs (EphA2, IL13Rα2, survivin), emulsified in Montanide-ISA-51, were administered subcutaneously adjacent to intramuscular injections of poly-ICLC every 3 weeks for 8 courses, followed by booster vaccines every 6 weeks. Primary endpoints were safety and T-cell responses against the GAA epitopes, assessed by enzyme-linked immunosorbent spot (ELISPOT) analysis. Treatment response was evaluated clinically and by magnetic resonance imaging. Twelve children were enrolled, 6 with glioblastoma, 5 with anaplastic astrocytoma, and one with malignant gliomatosis cerebri. No dose-limiting non-CNS toxicity was encountered. ELISPOT analysis, in ten children, showed GAA responses in 9: to IL13Rα2 in 4, EphA2 in 9, and survivin in 3. One child had presumed symptomatic pseudoprogression, discontinued vaccine therapy, and responded to subsequent treatment. One other child had a partial response that persisted throughout 2 years of vaccine therapy, and continues at >39 months. Median progression-free survival (PFS) from the start of vaccination was 4.1 months and median overall survival (OS) was 12.9 months. 6-month PFS and OS were 33 and 73 %, respectively. GAA peptide vaccination in children with recurrent malignant gliomas is generally well tolerated, and has preliminary evidence of immunological and modest clinical activity.
Collapse
|
76
|
Schmidt AE, Refaai MA, Blumberg N. Past, present and forecast of transfusion medicine: What has changed and what is expected to change? Presse Med 2016; 45:e253-72. [PMID: 27474234 DOI: 10.1016/j.lpm.2016.06.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Blood transfusion is the second most used medical procedures in health care systems worldwide. Over the last few decades, significant changes have been evolved in transfusion medicine practices. These changes were mainly needed to increase safety, efficacy, and availability of blood products as well as reduce recipients' unnecessary exposure to allogeneic blood. Blood products collection, processing, and storage as well as transfusion practices throughout all patient populations were the main stream of these changes. Health care systems across the world have adopted some or most of these changes to reduce transfusion risks, to improve overall patients' outcome, and to reduce health care costs. In this article, we are going to present and discuss some of these recent modifications and their impact on patients' safety.
Collapse
Affiliation(s)
- Amy E Schmidt
- University of Rochester medical center, department of pathology and laboratory medicine, 14642 Rochester, NY, USA
| | - Majed A Refaai
- University of Rochester medical center, department of pathology and laboratory medicine, 14642 Rochester, NY, USA
| | - Neil Blumberg
- University of Rochester medical center, department of pathology and laboratory medicine, 14642 Rochester, NY, USA.
| |
Collapse
|
77
|
Zeltsman M, Mayor M, Jones DR, Adusumilli PS. Surgical immune interventions for solid malignancies. Am J Surg 2016; 212:682-690.e5. [PMID: 27659157 DOI: 10.1016/j.amjsurg.2016.06.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 06/17/2016] [Accepted: 06/17/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND The purpose of this study was to systematically review clinically translatable immunotherapeutic agents that are delivered regionally for solid malignancies. DATA SOURCES PubMed and ClinicalTrials.gov were searched for published and registered clinical trials, respectively. The search yielded 334 relevant publications, of which 116 articles were included for review after exclusion criteria were applied. CONCLUSIONS There has been an increase in the regional administration of cell-based and viral vector-based clinical trials over the last 5 years. Surgical interventions have been developed for intrapleural, intracranial, intraperitoneal, and intratumoral routes of access to enhance the local delivery of these therapies. Multimodality therapies that combine regional immunotherapy with other local and systemic therapies are demonstrating continued growth as the field of immunotherapy continues to expand.
Collapse
Affiliation(s)
- Masha Zeltsman
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, USA
| | - Marissa Mayor
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, USA
| | - David R Jones
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, USA
| | - Prasad S Adusumilli
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, USA; Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, USA.
| |
Collapse
|
78
|
Müller I, Altherr D, Eyrich M, Flesch B, Friedmann KS, Ketter R, Oertel J, Schwarz EC, Technau A, Urbschat S, Eichler H. Tumor antigen-specific T cells for immune monitoring of dendritic cell-treated glioblastoma patients. Cytotherapy 2016; 18:1146-61. [PMID: 27424145 DOI: 10.1016/j.jcyt.2016.05.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 04/26/2016] [Accepted: 05/20/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND AIMS CD8(+) T cells are part of the adaptive immune system and, as such, are responsible for the elimination of tumor cells. Dendritic cells (DC) are professional antigen-presenting cells (APC) that activate CD8(+) T cells. Effector CD8(+) T cells in turn mediate the active immunotherapeutic response of DC vaccination against the aggressive glioblastoma (GBM). The lack of tumor response assays complicates the assessment of treatment success in GBM patients. METHODS A novel assay to identify specific cytotoxicity of activated T cells by APC was evaluated. Tumor antigen-pulsed DCs from HLA-A*02-positive GBM patients were cultivated to stimulate autologous cytotoxic T lymphocytes (CTL) over a 12-day culture period. To directly correlate antigen specificity and cytotoxic capacity, intracellular interferon (IFN)-γ fluorescence flow cytometry-based measurements were combined with anti-GBM tumor peptide dextramer staining. IFN-γ response was quantified by real-time polymerase chain reaction (PCR), and selected GBM genes were compared with healthy human brain cDNA by single specific primer PCR characterization. RESULTS Using CTL of GBM patients stimulated with GBM lysate-pulsed DCs increased IFN-γ messenger RNA levels, and intracellular IFN-γ protein expression was positively correlated with specificity against GBM antigens. Moreover, the GBM peptide-specific CD8(+) T-cell response correlated with specific GBM gene expression. Following DC vaccination, GBM patients showed 10-fold higher tumor-specific signals compared with unvaccinated GBM patients. DISCUSSION These data indicate that GBM tumor peptide-dextramer staining of CTL in combination with intracellular IFN-γ staining may be a useful tool to acquire information on whether a specific tumor antigen has the potential to induce an immune response in vivo.
Collapse
Affiliation(s)
- Isabelle Müller
- Institute of Clinical Hemostaseology and Transfusion Medicine, Saarland University Medical Center, Homburg, Germany.
| | - Dominik Altherr
- Institute of Clinical Hemostaseology and Transfusion Medicine, Saarland University Medical Center, Homburg, Germany
| | - Matthias Eyrich
- Stem Cell Laboratory, University Children's Hospital, University of Würzburg, Würzburg, Germany
| | - Brigitte Flesch
- Immungenetic/HLA, German Red Cross Blood Service, Bad Kreuznach, Germany
| | - Kim S Friedmann
- Biophysics, Center for Integrative Physiology and Molecular Medicine, Saarland University School of Medicine, Homburg, Germany
| | - Ralf Ketter
- Department of Neurosurgery, Saarland University Medical Center, Homburg, Germany
| | - Joachim Oertel
- Department of Neurosurgery, Saarland University Medical Center, Homburg, Germany
| | - Eva C Schwarz
- Biophysics, Center for Integrative Physiology and Molecular Medicine, Saarland University School of Medicine, Homburg, Germany
| | - Antje Technau
- Stem Cell Laboratory, University Children's Hospital, University of Würzburg, Würzburg, Germany
| | - Steffi Urbschat
- Department of Neurosurgery, Saarland University Medical Center, Homburg, Germany
| | - Hermann Eichler
- Institute of Clinical Hemostaseology and Transfusion Medicine, Saarland University Medical Center, Homburg, Germany
| |
Collapse
|
79
|
Chen K, Wang JM, Yuan R, Yi X, Li L, Gong W, Yang T, Li L, Su S. Tissue-resident dendritic cells and diseases involving dendritic cell malfunction. Int Immunopharmacol 2016; 34:1-15. [PMID: 26906720 PMCID: PMC4818737 DOI: 10.1016/j.intimp.2016.02.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/05/2016] [Indexed: 01/10/2023]
Abstract
Dendritic cells (DCs) control immune responses and are central to the development of immune memory and tolerance. DCs initiate and orchestrate immune responses in a manner that depends on signals they receive from microbes and cellular environment. Although DCs consist mainly of bone marrow-derived and resident populations, a third tissue-derived population resides the spleen and lymph nodes (LNs), different subsets of tissue-derived DCs have been identified in the blood, spleen, lymph nodes, skin, lung, liver, gut and kidney to maintain the tolerance and control immune responses. Tissue-resident DCs express different receptors for microbe-associated molecular patterns (MAMPs) and damage-associated molecular patterns (DAMPs), which were activated to promote the production of pro- or anti-inflammatory cytokines. Malfunction of DCs contributes to diseases such as autoimmunity, allergy, and cancer. It is therefore important to update the knowledge about resident DC subsets and diseases associated with DC malfunction.
Collapse
Affiliation(s)
- Keqiang Chen
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA; Laboratory of Inflammation Biology, Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061-0910, USA.
| | - Ji Ming Wang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA.
| | - Ruoxi Yuan
- Laboratory of Inflammation Biology, Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061-0910, USA
| | - Xiang Yi
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Liangzhu Li
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | - Wanghua Gong
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA; Basic Research Program, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Tianshu Yang
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Liwu Li
- Laboratory of Inflammation Biology, Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061-0910, USA
| | - Shaobo Su
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| |
Collapse
|
80
|
Pollack IF, Jakacki RI, Butterfield LH, Hamilton RL, Panigrahy A, Normolle DP, Connelly AK, Dibridge S, Mason G, Whiteside TL, Okada H. Immune responses and outcome after vaccination with glioma-associated antigen peptides and poly-ICLC in a pilot study for pediatric recurrent low-grade gliomas. Neuro Oncol 2016; 18:1157-68. [PMID: 26984745 DOI: 10.1093/neuonc/now026] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 01/29/2016] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Low-grade gliomas (LGGs) are the most common brain tumors of childhood. Although surgical resection is curative for well-circumscribed superficial lesions, tumors that are infiltrative or arise from deep structures are therapeutically challenging, and new treatment approaches are needed. Having identified a panel of glioma-associated antigens (GAAs) overexpressed in these tumors, we initiated a pilot trial of vaccinations with peptides for GAA epitopes in human leukocyte antigen-A2+ children with recurrent LGG that had progressed after at least 2 prior regimens. METHODS Peptide epitopes for 3 GAAs (EphA2, IL-13Rα2, and survivin) were emulsified in Montanide-ISA-51 and administered subcutaneously adjacent to intramuscular injections of polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose every 3 weeks for 8 courses, followed by booster vaccines every 6 weeks. Primary endpoints were safety and T-lymphocyte responses against GAA epitopes. Treatment response was evaluated clinically and by MRI. RESULTS Fourteen children were enrolled. Other than grade 3 urticaria in one child, no regimen-limiting toxicity was encountered. Vaccination induced immunoreactivity to at least one vaccine-targeted GAA in all 12 evaluable patients: to IL-13Rα2 in 3, EphA2 in 11, and survivin in 3. One child with a metastatic LGG had asymptomatic pseudoprogression noted 6 weeks after starting vaccination, followed by dramatic disease regression with >75% shrinkage of primary tumor and regression of metastatic disease, persisting >57 months. Three other children had sustained partial responses, lasting >10, >31, and >45 months, and one had a transient response. CONCLUSIONS GAA peptide vaccination in children with recurrent LGGs is generally well tolerated, with preliminary evidence of immunological and clinical activity.
Collapse
Affiliation(s)
- Ian F Pollack
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., H.O.); Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania (R.I.J., A.K.C., S.D., G.M.); Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (R.L.H., T.L.W.); Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B.), Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B., H.O.), Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania (A.P.), Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B., T.L.W.), University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., R.I.J., A.P., A.K.C., S.D., G.M.); University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., R.I.J., L.H.B., R.L.H., D.P.N., G.M., T.L.W., H.O.); Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania (D.P.N.); Department of Neurosurgery, University of CaliforniaSan Francisco, San Francisco, California (H.O.)
| | - Regina I Jakacki
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., H.O.); Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania (R.I.J., A.K.C., S.D., G.M.); Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (R.L.H., T.L.W.); Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B.), Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B., H.O.), Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania (A.P.), Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B., T.L.W.), University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., R.I.J., A.P., A.K.C., S.D., G.M.); University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., R.I.J., L.H.B., R.L.H., D.P.N., G.M., T.L.W., H.O.); Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania (D.P.N.); Department of Neurosurgery, University of CaliforniaSan Francisco, San Francisco, California (H.O.)
| | - Lisa H Butterfield
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., H.O.); Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania (R.I.J., A.K.C., S.D., G.M.); Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (R.L.H., T.L.W.); Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B.), Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B., H.O.), Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania (A.P.), Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B., T.L.W.), University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., R.I.J., A.P., A.K.C., S.D., G.M.); University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., R.I.J., L.H.B., R.L.H., D.P.N., G.M., T.L.W., H.O.); Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania (D.P.N.); Department of Neurosurgery, University of CaliforniaSan Francisco, San Francisco, California (H.O.)
| | - Ronald L Hamilton
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., H.O.); Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania (R.I.J., A.K.C., S.D., G.M.); Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (R.L.H., T.L.W.); Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B.), Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B., H.O.), Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania (A.P.), Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B., T.L.W.), University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., R.I.J., A.P., A.K.C., S.D., G.M.); University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., R.I.J., L.H.B., R.L.H., D.P.N., G.M., T.L.W., H.O.); Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania (D.P.N.); Department of Neurosurgery, University of CaliforniaSan Francisco, San Francisco, California (H.O.)
| | - Ashok Panigrahy
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., H.O.); Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania (R.I.J., A.K.C., S.D., G.M.); Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (R.L.H., T.L.W.); Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B.), Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B., H.O.), Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania (A.P.), Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B., T.L.W.), University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., R.I.J., A.P., A.K.C., S.D., G.M.); University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., R.I.J., L.H.B., R.L.H., D.P.N., G.M., T.L.W., H.O.); Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania (D.P.N.); Department of Neurosurgery, University of CaliforniaSan Francisco, San Francisco, California (H.O.)
| | - Daniel P Normolle
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., H.O.); Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania (R.I.J., A.K.C., S.D., G.M.); Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (R.L.H., T.L.W.); Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B.), Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B., H.O.), Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania (A.P.), Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B., T.L.W.), University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., R.I.J., A.P., A.K.C., S.D., G.M.); University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., R.I.J., L.H.B., R.L.H., D.P.N., G.M., T.L.W., H.O.); Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania (D.P.N.); Department of Neurosurgery, University of CaliforniaSan Francisco, San Francisco, California (H.O.)
| | - Angela K Connelly
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., H.O.); Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania (R.I.J., A.K.C., S.D., G.M.); Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (R.L.H., T.L.W.); Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B.), Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B., H.O.), Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania (A.P.), Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B., T.L.W.), University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., R.I.J., A.P., A.K.C., S.D., G.M.); University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., R.I.J., L.H.B., R.L.H., D.P.N., G.M., T.L.W., H.O.); Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania (D.P.N.); Department of Neurosurgery, University of CaliforniaSan Francisco, San Francisco, California (H.O.)
| | - Sharon Dibridge
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., H.O.); Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania (R.I.J., A.K.C., S.D., G.M.); Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (R.L.H., T.L.W.); Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B.), Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B., H.O.), Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania (A.P.), Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B., T.L.W.), University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., R.I.J., A.P., A.K.C., S.D., G.M.); University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., R.I.J., L.H.B., R.L.H., D.P.N., G.M., T.L.W., H.O.); Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania (D.P.N.); Department of Neurosurgery, University of CaliforniaSan Francisco, San Francisco, California (H.O.)
| | - Gary Mason
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., H.O.); Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania (R.I.J., A.K.C., S.D., G.M.); Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (R.L.H., T.L.W.); Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B.), Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B., H.O.), Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania (A.P.), Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B., T.L.W.), University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., R.I.J., A.P., A.K.C., S.D., G.M.); University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., R.I.J., L.H.B., R.L.H., D.P.N., G.M., T.L.W., H.O.); Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania (D.P.N.); Department of Neurosurgery, University of CaliforniaSan Francisco, San Francisco, California (H.O.)
| | - Theresa L Whiteside
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., H.O.); Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania (R.I.J., A.K.C., S.D., G.M.); Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (R.L.H., T.L.W.); Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B.), Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B., H.O.), Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania (A.P.), Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B., T.L.W.), University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., R.I.J., A.P., A.K.C., S.D., G.M.); University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., R.I.J., L.H.B., R.L.H., D.P.N., G.M., T.L.W., H.O.); Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania (D.P.N.); Department of Neurosurgery, University of CaliforniaSan Francisco, San Francisco, California (H.O.)
| | - Hideho Okada
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., H.O.); Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania (R.I.J., A.K.C., S.D., G.M.); Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (R.L.H., T.L.W.); Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B.), Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B., H.O.), Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania (A.P.), Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania (L.H.B., T.L.W.), University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., R.I.J., A.P., A.K.C., S.D., G.M.); University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania (I.F.P., R.I.J., L.H.B., R.L.H., D.P.N., G.M., T.L.W., H.O.); Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania (D.P.N.); Department of Neurosurgery, University of CaliforniaSan Francisco, San Francisco, California (H.O.)
| |
Collapse
|
81
|
Vandenberk L, Belmans J, Van Woensel M, Riva M, Van Gool SW. Exploiting the Immunogenic Potential of Cancer Cells for Improved Dendritic Cell Vaccines. Front Immunol 2016; 6:663. [PMID: 26834740 PMCID: PMC4712296 DOI: 10.3389/fimmu.2015.00663] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 12/26/2015] [Indexed: 12/31/2022] Open
Abstract
Cancer immunotherapy is currently the hottest topic in the oncology field, owing predominantly to the discovery of immune checkpoint blockers. These promising antibodies and their attractive combinatorial features have initiated the revival of other effective immunotherapies, such as dendritic cell (DC) vaccinations. Although DC-based immunotherapy can induce objective clinical and immunological responses in several tumor types, the immunogenic potential of this monotherapy is still considered suboptimal. Hence, focus should be directed on potentiating its immunogenicity by making step-by-step protocol innovations to obtain next-generation Th1-driving DC vaccines. We review some of the latest developments in the DC vaccination field, with a special emphasis on strategies that are applied to obtain a highly immunogenic tumor cell cargo to load and to activate the DCs. To this end, we discuss the effects of three immunogenic treatment modalities (ultraviolet light, oxidizing treatments, and heat shock) and five potent inducers of immunogenic cell death [radiotherapy, shikonin, high-hydrostatic pressure, oncolytic viruses, and (hypericin-based) photodynamic therapy] on DC biology and their application in DC-based immunotherapy in preclinical as well as clinical settings.
Collapse
Affiliation(s)
- Lien Vandenberk
- Laboratory of Pediatric Immunology, Department of Immunology and Microbiology, KU Leuven University of Leuven , Leuven , Belgium
| | - Jochen Belmans
- Laboratory of Pediatric Immunology, Department of Immunology and Microbiology, KU Leuven University of Leuven , Leuven , Belgium
| | - Matthias Van Woensel
- Laboratory of Experimental and Neuroanatomy, Department of Neurosciences, KU Leuven University of Leuven, Leuven, Belgium; Laboratory of Pharmaceutics and Biopharmaceutics, Université Libre de Bruxelles, Brussels, Belgium
| | - Matteo Riva
- Laboratory of Pediatric Immunology, Department of Immunology and Microbiology, KU Leuven University of Leuven, Leuven, Belgium; Department of Neurosurgery, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | - Stefaan W Van Gool
- Laboratory of Pediatric Immunology, Department of Immunology and Microbiology, KU Leuven University of Leuven, Leuven, Belgium; Kinderklinik, RWTH, Aachen, Germany; Immunologic-Oncologic Centre Cologne (IOZK), Köln, Germany
| |
Collapse
|
82
|
Stress Response Leading to Resistance in Glioblastoma-The Need for Innovative Radiotherapy (iRT) Concepts. Cancers (Basel) 2016; 8:cancers8010015. [PMID: 26771644 PMCID: PMC4728462 DOI: 10.3390/cancers8010015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 12/23/2015] [Accepted: 01/08/2016] [Indexed: 11/16/2022] Open
Abstract
Glioblastoma (GBM) is the most common and most aggressive malignant primary brain tumor in adults. In spite of multimodal therapy concepts, consisting of surgery, radiotherapy and chemotherapy, the median survival, merely 15–18 months, is still poor. Mechanisms for resistance of GBM to radio(chemo)therapy are not fully understood yet and due to the genetic heterogeneity within the tumor including radiation-resistant tumor stem cells, there are several factors leading to therapy failure. Recent research revealed that, hypoxia during radiation and miRNAs may adversely affect the therapeutic response to radiotherapy. Further molecular alterations and prognostic markers like the DNA-repair protein O6-methylguanine-DNA methyltransferase (MGMT), anti-apoptotic molecular chaperones, and/or the activity of aldehyde dehydrogenase 1 (ALDH1) have also been identified to play a role in the sensitivity to cytostatic agents. Latest approaches in the field of radiotherapy to use particle irradiation or dose escalation strategies including modern molecular imaging, however, need further evaluation with regard to long-term outcome. In this review we focus on current information about the mechanisms and markers that mediate resistance to radio(chemo)therapy, and discuss the opportunities of Innovative Radiotherapy (iRT) concepts to improve treatment options for GBM patients.
Collapse
|
83
|
Finocchiaro G, Pellegatta S. Immunotherapy with dendritic cells loaded with glioblastoma stem cells: from preclinical to clinical studies. Cancer Immunol Immunother 2016; 65:101-9. [PMID: 26377689 PMCID: PMC11029491 DOI: 10.1007/s00262-015-1754-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 08/23/2015] [Indexed: 01/18/2023]
Abstract
Different approaches have been explored to raise effective antitumor responses against glioblastoma (GBM), the deadliest of primary brain tumors. In many clinical studies, cancer vaccines have been based on dendritic cells (DCs) loaded with peptides, representing one or more specific tumor antigens or whole lysates as a source of multiple antigens. Randomized clinical trials using DCs are ongoing, and results of efficacy are not yet available. Such strategies are feasible and safe; however, immune-suppressive microenvironment, absence of appropriate specific epitopes to target, and cancer immunoediting can limit their efficacy. The aim of this review is to describe how the definition of novel and more specific targets may increase considerably the possibility of successful DC immunotherapy. By proposing to target glioblastoma stem-like cells (GSCs), the immune response will be pointed to eradicating factors and pathways highly relevant to GBM biology. Preclinical observations on efficacy, and preliminary results of immunotherapy trials, encourage exploring the clinical efficacy of DC immunotherapy in GBM patients using high-purity, GSC-loaded DC vaccines.
Collapse
Affiliation(s)
- Gaetano Finocchiaro
- Unit of Molecular Neuro-Oncology, Fondazione I.R.C.C.S. Istituto Neurologico C. Besta, Via Celoria 11, 20133, Milan, Italy.
| | - Serena Pellegatta
- Unit of Molecular Neuro-Oncology, Fondazione I.R.C.C.S. Istituto Neurologico C. Besta, Via Celoria 11, 20133, Milan, Italy.
| |
Collapse
|
84
|
Immunomonitoring in glioma immunotherapy: current status and future perspectives. J Neurooncol 2015; 127:1-13. [PMID: 26638171 DOI: 10.1007/s11060-015-2018-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Accepted: 11/25/2015] [Indexed: 12/28/2022]
Abstract
Given the continued poor clinical outcomes and refractory nature of glioblastoma multiforme to traditional interventions, immunotherapy is gaining traction due to its potential for specific tumor-targeting and long-term antitumor protective surveillance. Currently, development of glioma immunotherapy relies on overall survival as an endpoint in clinical trials. However, the identification of surrogate immunologic biomarkers can accelerate the development of successful immunotherapeutic strategies. Immunomonitoring techniques possess the potential to elucidate immunological mechanisms of antitumor responses, monitor disease progression, evaluate therapeutic effect, identify candidates for immunotherapy, and serve as prognostic markers of clinical outcome. Current immunomonitoring assays assess delayed-type hypersensitivity, T cell proliferation, cytotoxic T-lymphocyte function, cytokine secretion profiles, antibody titers, and lymphocyte phenotypes. Yet, no single immunomonitoring technique can reliably predict outcomes, relegating immunological markers to exploratory endpoints. In response, the most recent immunomonitoring assays are incorporating emerging technologies and novel analysis techniques to approach the goal of identifying a competent immunological biomarker which predicts therapy responsiveness and clinical outcome. This review addresses the current status of immunomonitoring in glioma vaccine clinical trials with emphasis on correlations with clinical response.
Collapse
|
85
|
|
86
|
Polyzoidis S, Ashkan K. DCVax®-L--developed by Northwest Biotherapeutics. Hum Vaccin Immunother 2015; 10:3139-45. [PMID: 25483653 DOI: 10.4161/hv.29276] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Dendritic cell (DC) immunotherapy is emerging as a potential addition to the standard of care in the treatment of glioblastoma multiforme (GBM). In the last decade or so various research groups have conducted phase I and II trials of DC-immunotherapy on patients with newly diagnosed (ND) and recurrent GBM and other high-grade gliomas in an attempt to improve the poor prognosis. Results show an increase in overall survival (OS), while vaccination-related side effects are invariably mild. Northwest Biotherapeutics, Inc., Bethesda, Maryland, U.S.A. (NWBT) developed the DCVax®-L vaccine as an adjunct to the treatment of GBM. It is currently under evaluation in a phase III trial in patients with ND-GBM, which is the only ongoing trial of its kind. In this review current data and perspectives of this product are examined.
Collapse
Key Words
- BBB, blood brain barrier
- CNS, central nervous system
- CTL, cytotoxic T-lymphocyte
- DC, dendritic cell
- DCVax®-L
- DTH, delayed tissue hypersensitivity
- EORTC, European Organization for Research and Treatment of Cancer
- FDA, Food and Drug Administration
- GBM, glioblastoma multiforme
- GM-CSF, granulocyte-macrophage colony-stimulating factor
- HGG, high-grade glioma
- IL-4, interleukin-4
- IMP, investigational medicinal product
- MHRA, Medicines and Healthcare products Regulatory Agency
- MRI, magnetic resonance imaging
- ND, newly diagnosed
- NIHR, National Institute for Health Research
- NWBT, Northwest Biotherapeutics Inc.
- OS, overall survival
- PEI, Paul-Ehrlich-Institute
- PFS, progression-free survival
- TAAs, tumor-associated antigens
- UCLA, University of California, Los Angeles, U.S.A., United States of America
- dendritic cells
- glioblastoma multiforme
- immunotherapy
- overall survival
- side effects
- vaccine
Collapse
Affiliation(s)
- Stavros Polyzoidis
- a Department of Neurosurgery; King's College Hospital; King's College ; London , UK
| | | |
Collapse
|
87
|
Vandenberk L, Garg AD, Verschuere T, Koks C, Belmans J, Beullens M, Agostinis P, De Vleeschouwer S, Van Gool SW. Irradiation of necrotic cancer cells, employed for pulsing dendritic cells (DCs), potentiates DC vaccine-induced antitumor immunity against high-grade glioma. Oncoimmunology 2015; 5:e1083669. [PMID: 27057467 PMCID: PMC4801426 DOI: 10.1080/2162402x.2015.1083669] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 07/31/2015] [Accepted: 08/11/2015] [Indexed: 12/05/2022] Open
Abstract
Dendritic cell (DC)-based immunotherapy has yielded promising results against high-grade glioma (HGG). However, the efficacy of DC vaccines is abated by HGG-induced immunosuppression and lack of attention toward the immunogenicity of the tumor lysate/cells used for pulsing DCs. A literature analysis of DC vaccination clinical trials in HGG patients delineated the following two most predominantly applied methods for tumor lysate preparation: freeze-thaw (FT)-induced necrosis or FT-necrosis followed by X-ray irradiation. However, from the available clinical evidence, it is unclear which of both methodologies has superior immunogenic potential. Using an orthotopic HGG murine model (GL261-C57BL/6), we observed that prophylactic vaccination with DCs pulsed with irradiated FT-necrotic cells (compared to FT-necrotic cells only) prolonged overall survival by increasing tumor rejection in glioma-challenged mice. This was associated, both in prophylactic and curative vaccination setups, with an increase in brain-infiltrating Th1 cells and cytotoxic T lymphocytes (CTL), paralleled by a reduced accumulation of regulatory T cells, tumor-associated macrophages (TAM) and myeloid-derived suppressor cells (MDSC). Further analysis showed that irradiation treatment of FT-necrotic cells considerably increased the levels of carbonylated proteins — a surrogate-marker of oxidation-associated molecular patterns (OAMPs). Through further application of antioxidants and hydrogen peroxide, we found a striking correlation between the amount of lysate-associated protein carbonylation/OAMPs and DC vaccine-mediated tumor rejection capacity thereby suggesting for the first time a role for protein carbonylation/OAMPs in at least partially mediating antitumor immunity. Together, these data strongly advocate the use of protein oxidation-inducing modalities like irradiation for increasing the immunogenicity of tumor lysate/cells used for pulsing DC vaccines.
Collapse
Affiliation(s)
- Lien Vandenberk
- KU Leuven - University of Leuven, Department of Microbiology and Immunology, Laboratory of Pediatric Immunology , Leuven, Belgium
| | - Abhishek D Garg
- KU Leuven - University of Leuven, Department of Cellular and Molecular Medicine, Laboratory of Cell Death Research and Therapy , Leuven, Belgium
| | - Tina Verschuere
- KU Leuven - University of Leuven, Department of Microbiology and Immunology, Laboratory of Pediatric Immunology , Leuven, Belgium
| | - Carolien Koks
- KU Leuven - University of Leuven, Department of Microbiology and Immunology, Laboratory of Pediatric Immunology , Leuven, Belgium
| | - Jochen Belmans
- KU Leuven - University of Leuven, Department of Microbiology and Immunology, Laboratory of Pediatric Immunology , Leuven, Belgium
| | - Monique Beullens
- KU Leuven - University of Leuven, Department of Cellular and Molecular Medicine, Laboratory of Biosignaling and Therapeutics , Leuven, Belgium
| | - Patrizia Agostinis
- KU Leuven - University of Leuven, Department of Cellular and Molecular Medicine, Laboratory of Cell Death Research and Therapy , Leuven, Belgium
| | - Steven De Vleeschouwer
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurosurgery and Neuroanatomy , Leuven, Belgium
| | - Stefaan W Van Gool
- KU Leuven - University of Leuven, Department of Microbiology and Immunology, Laboratory of Pediatric Immunology , Leuven, Belgium
| |
Collapse
|
88
|
Sakai K, Shimodaira S, Maejima S, Udagawa N, Sano K, Higuchi Y, Koya T, Ochiai T, Koide M, Uehara S, Nakamura M, Sugiyama H, Yonemitsu Y, Okamoto M, Hongo K. Dendritic cell-based immunotherapy targeting Wilms' tumor 1 in patients with recurrent malignant glioma. J Neurosurg 2015; 123:989-97. [PMID: 26252465 DOI: 10.3171/2015.1.jns141554] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECT Dendritic cell (DC)-based vaccination is considered a potentially effective therapy against advanced cancer. The authors conducted a Phase I study to investigate the safety and immunomonitoring of Wilms' tumor 1 (WT1)-pulsed DC vaccination therapy for patients with relapsed malignant glioma. METHODS WT1-pulsed and/or autologous tumor lysate-pulsed DC vaccination therapy was performed in patients with relapsed malignant gliomas. Approximately 1 × 10(7) to 2 × 10(7) pulsed DCs loaded with WT1 peptide antigen and/or tumor lysate were intradermally injected into the axillary areas with OK-432, a streptococcal preparation, at 2-week intervals for at least 5-7 sessions (1 course) during an individual chemotherapy regimen. RESULTS Ten patients (3 men, 7 women; age range 24-64 years [median 39 years]) with the following tumors were enrolled: glioblastoma (6), anaplastic astrocytoma (2), anaplastic oligoastrocytoma (1), and anaplastic oligodendroglioma (1). Modified WT1 peptide-pulsed DC vaccine was administered to 7 patients, tumor lysate-pulsed DC vaccine to 2 patients, and both tumor lysate-pulsed and WT1-pulsed DC vaccine to 1 patient. The clinical response was stable disease in 5 patients with WT1-pulsed DC vaccination. In 2 of 5 patients with stable disease, neurological findings improved, and MR images showed tumor shrinkage. No serious adverse events occurred except Grade 1-2 erythema at the injection sites. WT1 tetramer analysis detected WT1-reactive cytotoxic T cells after vaccination in patients treated with WT1-pulsed therapy. Positivity for skin reaction at the injection sites was 80% (8 of 10 patients) after the first session, and positivity remained for these 8 patients after the final session. CONCLUSIONS This study of WT1-pulsed DC vaccination therapy demonstrated safety, immunogenicity, and feasibility in the management of relapsed malignant gliomas.
Collapse
Affiliation(s)
- Keiichi Sakai
- Department of Neurosurgery, Shinshu University School of Medicine;,Department of Neurosurgery, National Hospital Organization, Shinshu Ueda Medical Center, Ueda, Nagano, Japan
| | | | | | | | - Kenji Sano
- Department of Laboratory, Shinshu University Hospital, Matsumoto
| | - Yumiko Higuchi
- Center for Advanced Cellular Therapy, Shinshu University Hospital, Matsumoto
| | - Terutsugu Koya
- Center for Advanced Cellular Therapy, Shinshu University Hospital, Matsumoto
| | | | | | - Shunsuke Uehara
- Department of Biochemistry, Matsumoto Dental University Hospital, Shiojiri
| | - Midori Nakamura
- Department of Biochemistry, Matsumoto Dental University Hospital, Shiojiri
| | - Haruo Sugiyama
- Department of Functional Diagnostic Science, Graduate School of Medicine, Osaka University, Osaka
| | - Yoshikazu Yonemitsu
- R&D Laboratory for Innovative Biotherapeutics, Graduate School of Pharmaceutical Sciences, Kyushu University, Higashi-ku, Fukuoka
| | - Masato Okamoto
- Department of Advanced Immunotherapeutics, Kitasato University School of Pharmacy, Minato-ku, Tokyo; and
| | - Kazuhiro Hongo
- Department of Neurosurgery, Shinshu University School of Medicine
| |
Collapse
|
89
|
Fecci PE, Heimberger AB, Sampson JH. Immunotherapy for primary brain tumors: no longer a matter of privilege. Clin Cancer Res 2015; 20:5620-9. [PMID: 25398845 DOI: 10.1158/1078-0432.ccr-14-0832] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Immunotherapy for cancer continues to gain both momentum and legitimacy as a rational mode of therapy and a vital treatment component in the emerging era of personalized medicine. Gliomas, and their most malignant form, glioblastoma, remain as a particularly devastating solid tumor for which standard treatment options proffer only modest efficacy and target specificity. Immunotherapy would seem a well-suited choice to address such deficiencies given both the modest inherent immunogenicity of gliomas and the strong desire for treatment specificity within the confines of the toxicity-averse normal brain. This review highlights the caveats and challenges to immunotherapy for primary brain tumors, as well as reviewing modalities that are currently used or are undergoing active investigation. Tumor immunosuppressive countermeasures, peculiarities of central nervous system immune access, and opportunities for rational treatment design are discussed.
Collapse
Affiliation(s)
- Peter E Fecci
- Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John H Sampson
- Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina.
| |
Collapse
|
90
|
Van Gool SW. Brain Tumor Immunotherapy: What have We Learned so Far? Front Oncol 2015; 5:98. [PMID: 26137448 PMCID: PMC4470276 DOI: 10.3389/fonc.2015.00098] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 04/13/2015] [Indexed: 12/17/2022] Open
Abstract
High grade glioma is a rare brain cancer, incurable in spite of modern neurosurgery, radiotherapy, and chemotherapy. Novel approaches are in research, and immunotherapy emerges as a promising strategy. Clinical experiences with active specific immunotherapy demonstrate feasibility, safety and most importantly, but incompletely understood, prolonged long-term survival in a fraction of the patients. In relapsed patients, we developed an immunotherapy schedule and we categorized patients into clinically defined risk profiles. We learned how to combine immunotherapy with standard multimodal treatment strategies for newly diagnosed glioblastoma multiforme patients. The developmental program allows further improvements related to newest scientific insights. Finally, we developed a mode of care within academic centers to organize cell-based therapies for experimental clinical trials in a large number of patients.
Collapse
|
91
|
Hashimoto N, Tsuboi A, Kagawa N, Chiba Y, Izumoto S, Kinoshita M, Kijima N, Oka Y, Morimoto S, Nakajima H, Morita S, Sakamoto J, Nishida S, Hosen N, Oji Y, Arita N, Yoshimine T, Sugiyama H. Wilms tumor 1 peptide vaccination combined with temozolomide against newly diagnosed glioblastoma: safety and impact on immunological response. Cancer Immunol Immunother 2015; 64:707-16. [PMID: 25772149 PMCID: PMC11028974 DOI: 10.1007/s00262-015-1674-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 02/25/2015] [Indexed: 11/28/2022]
Abstract
To investigate the safety of combined Wilms tumor 1 peptide vaccination and temozolomide treatment of glioblastoma, a phase I clinical trial was designed. Seven patients with histological diagnosis of glioblastoma underwent concurrent radiotherapy and temozolomide therapy. Patients first received Wilms tumor 1 peptide vaccination 1 week after the end of combined concurrent radio/temozolomide therapy, and administration was continued once per week for 7 weeks. Temozolomide maintenance was started and performed for up to 24 cycles, and the observation period for safety encompassed 6 weeks from the first administration of maintenance temozolomide. All patients showed good tolerability during the observation period. Skin disorders, such as grade 1/2 injection-site reactions, were observed in all seven patients. Although grade 3 lymphocytopenia potentially due to concurrent radio/temozolomide therapy was observed in five patients (71.4 %), no other grade 3/4 hematological or neurological toxicities were observed. No autoimmune reactions were observed. All patients are still alive, and six are on Wilms tumor 1 peptide vaccination without progression, yielding a progression-free survival from histological diagnosis of 5.2-49.1 months. Wilms tumor 1 peptide vaccination was stopped in one patient after 12 injections by the patient's request. The safety profile of the combined Wilms tumor 1 peptide vaccination and temozolomide therapy approach for treating glioblastoma was confirmed.
Collapse
Affiliation(s)
- Naoya Hashimoto
- Department of Neurosurgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Enhancement of the antigen-specific cytotoxic T lymphocyte-inducing ability in the PMDC11 leukemic plasmacytoid dendritic cell line via lentiviral vector-mediated transduction of the caTLR4 gene. Mol Med Rep 2015; 12:2443-50. [PMID: 25936433 PMCID: PMC4464268 DOI: 10.3892/mmr.2015.3685] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 02/03/2015] [Indexed: 12/16/2022] Open
Abstract
The aim of the present study was to enhance the efficiency of leukemia immunotherapy by increasing the antigen-specific cytotoxic T lymphocyte-inducing ability of leukemia cells. The leukemic plasmacytoid dendritic cell line PMDC05 containing the HLA-A02/24 antigen, which was previously established in our laboratory (Laboratory of Hematology and Oncology, Graduate School of Health Sciences, Niigata University, Niigata, Japan), was used in the present study. It exhibited higher expression levels of CD80 following transduction with lentiviruses encoding the CD80 gene. This CD80-expressing PMDC05 was named PMDC11. In order to establish a more potent antigen-presenting cell for cellular immunotherapy of tumors or severe infections, PMDC11 cells were transduced with a constitutively active (ca) toll-like receptor 4 (TLR4) gene using the Tet-On system (caTLR4-PMDC11). CD8+ T cells from healthy donors with HLA-A02 were co-cultured with mutant WT1 peptide-pulsed PMDC11, lipopolysaccharide (LPS)-stimulated PMDC11 or caTLR4-PMDC11 cells. Interleukin (IL)-2 (50 IU/ml) and IL-7 (10 ng/ml) were added on day three of culture. Priming with mutant WT1 peptide-pulsed PMDC11, LPS-stimulated PMDC11 or caTLR4-PMDC11 cells was conducted once per week and two thirds of the IL-2/IL-7 containing medium was replenished every 3–4 days. Immediately prior to the priming with these various PMDC11 cells, the cultured cells were analyzed for the secretion of interferon (IFN)-γ in addition to the percentage and number of CD8+/WT1 tetramer+ T cells using flow cytometry. caTLR4-PMDC11 cells were observed to possess greater antigen-presenting abilities compared with those of PMDC11 or LPS-stimulated PMDC11 cells in a mixed leukocyte culture. CD8 T cells positive for the WT1 tetramer were generated following 3–4 weeks of culture and CD8+/WT1 tetramer+ T cells were markedly increased in caTLR4-PMDC11-primed CD8+ T cell culture compared with PMDC11 or LPS-stimulated PMDC11-primed CD8+ T cell culture. These CD8+ T cells co-cultured with caTLR4-PMDC11 cells were demonstrated to secrete IFN-γ and to be cytotoxic to WT1-expressing target cells. These data suggested that the antigen-specific cytotoxic T lymphocyte (CTL)-inducing ability of PMDC11 was potentiated via transduction of the caTLR4 gene. The present study also suggested that caTLR4-PMDC11 cells may be applied as potent antigen-presenting cells for generating antigen-specific CTLs in adoptive cellular immunotherapy against tumors and severe viral infections.
Collapse
|
93
|
Mohammadi A, Mehrzad J, Mahmoudi M, Schneider M, Haghparast A. Effect of culture and maturation on human monocyte-derived dendritic cell surface markers, necrosis and antigen binding. Biotech Histochem 2015; 90:445-52. [DOI: 10.3109/10520295.2015.1017536] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
|
94
|
Glioblastoma stem cells and stem cell-targeting immunotherapies. J Neurooncol 2015; 123:449-57. [DOI: 10.1007/s11060-015-1729-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Accepted: 02/01/2015] [Indexed: 01/16/2023]
|
95
|
Rhun EL, Taillibert S, Chamberlain MC. The future of high-grade glioma: Where we are and where are we going. Surg Neurol Int 2015; 6:S9-S44. [PMID: 25722939 PMCID: PMC4338495 DOI: 10.4103/2152-7806.151331] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 10/15/2014] [Indexed: 01/12/2023] Open
Abstract
High-grade glioma (HGG) are optimally treated with maximum safe surgery, followed by radiotherapy (RT) and/or systemic chemotherapy (CT). Recently, the treatment of newly diagnosed anaplastic glioma (AG) has changed, particularly in patients with 1p19q codeleted tumors. Results of trials currenlty ongoing are likely to determine the best standard of care for patients with noncodeleted AG tumors. Trials in AG illustrate the importance of molecular characterization, which are germane to both prognosis and treatment. In contrast, efforts to improve the current standard of care of newly diagnosed glioblastoma (GB) with, for example, the addition of bevacizumab (BEV), have been largely disappointing and furthermore molecular characterization has not changed therapy except in elderly patients. Novel approaches, such as vaccine-based immunotherapy, for newly diagnosed GB are currently being pursued in multiple clinical trials. Recurrent disease, an event inevitable in nearly all patients with HGG, continues to be a challenge. Both recurrent GB and AG are managed in similar manner and when feasible re-resection is often suggested notwithstanding limited data to suggest benefit from repeat surgery. Occassional patients may be candidates for re-irradiation but again there is a paucity of data to commend this therapy and only a minority of selected patients are eligible for this approach. Consequently systemic therapy continues to be the most often utilized treatment in recurrent HGG. Choice of therapy, however, varies and revolves around re-challenge with temozolomide (TMZ), use of a nitrosourea (most often lomustine; CCNU) or BEV, the most frequently used angiogenic inhibitor. Nevertheless, no clear standard recommendation regarding the prefered agent or combination of agents is avaliable. Prognosis after progression of a HGG remains poor, with an unmet need to improve therapy.
Collapse
Affiliation(s)
- Emilie Le Rhun
- Department of Neuro-oncology, Roger Salengro Hospital, University Hospital, Lille, and Neurology, Department of Medical Oncology, Oscar Lambret Center, Lille, France, Inserm U-1192, Laboratoire de Protéomique, Réponse Inflammatoire, Spectrométrie de Masse (PRISM), Lille 1 University, Villeneuve D’Ascq, France
| | - Sophie Taillibert
- Neurology, Mazarin and Radiation Oncology, Pitié Salpétrière Hospital, University Pierre et Marie Curie, Paris VI, Paris, France
| | - Marc C. Chamberlain
- Department of Neurology and Neurological Surgery, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
96
|
Polyzoidis S, Tuazon J, Brazil L, Beaney R, Al-Sarraj ST, Doey L, Logan J, Hurwitz V, Jarosz J, Bhangoo R, Gullan R, Mijovic A, Richardson M, Farzaneh F, Ashkan K. Active dendritic cell immunotherapy for glioblastoma: Current status and challenges. Br J Neurosurg 2014; 29:197-205. [PMID: 25541743 DOI: 10.3109/02688697.2014.994473] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Dendritic cell (DC) immunotherapy is developing as a promising treatment modality for patients with glioblastoma multiforme (GBM). The aim of this article is to review the data from clinical trials and prospective studies evaluating the safety and efficacy of DC vaccines for newly diagnosed (ND)- and recurrent (Rec)-GBM and for other high-grade gliomas (HGGs). By searching all major databases we identified and reviewed twenty-two (n=22) such studies, twenty (n=20) of which were phase I and II trials, one was a pilot study towards a phase I/II trial and one was a prospective study. GBM patients were exclusively recruited in 12/22 studies, while 10/22 studies enrolled patients with any diagnosis of a HGG. In 7/22 studies GBM was newly diagnosed. In the vast majority of studies the vaccine was injected subcutaneously or intradermally and consisted of mature DCs pulsed with tumour lysate or peptides. Median overall survival ranged between 16.0 and 38.4 months for ND-GBM and between 9.6 and 35.9 months for Rec-GBM. Vaccine-related side effects were in general mild (grade I and II), with serious adverse events (grade III, IV and V) reported only rarely. DC immunotherapy therefore appears to have the potential to increase the overall survival in patients with HGG, with an acceptable side effect profile. The findings will require confirmation by the ongoing and future phase III trials.
Collapse
|
97
|
Yamanaka R, Hayano A. Experiences and expectations for glioma immunotherapeutic approaches. Front Oncol 2014; 4:355. [PMID: 25540770 PMCID: PMC4261702 DOI: 10.3389/fonc.2014.00355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 11/26/2014] [Indexed: 01/22/2023] Open
Affiliation(s)
- Ryuya Yamanaka
- Graduate School for Health Care Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Azusa Hayano
- Graduate School for Health Care Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
98
|
Rossmeisl JH. New treatment modalities for brain tumors in dogs and cats. Vet Clin North Am Small Anim Pract 2014; 44:1013-38. [PMID: 25441624 DOI: 10.1016/j.cvsm.2014.07.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Despite advancements in standard therapies, intracranial tumors remain a significant source of morbidity and mortality in veterinary and human medicine. Several newer approaches are gaining more widespread acceptance or are currently being prepared for translation from experimental to routine therapeutic use. Clinical trials in dogs with spontaneous brain tumors have contributed to the development and human translation of several novel therapeutic brain tumor approaches.
Collapse
Affiliation(s)
- John H Rossmeisl
- Neurology and Neurosurgery, Department of Small Animal Clinical Sciences, VA-MD Regional College of Veterinary Medicine, Virginia Tech, 215 Duckpond Drive, Mail Code 0442, Blacksburg, VA 24061, USA.
| |
Collapse
|
99
|
Galluzzi L, Senovilla L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial watch: Dendritic cell-based interventions for cancer therapy. Oncoimmunology 2014; 1:1111-1134. [PMID: 23170259 PMCID: PMC3494625 DOI: 10.4161/onci.21494] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Dendritic cells (DCs) occupy a central position in the immune system, orchestrating a wide repertoire of responses that span from the development of self-tolerance to the elicitation of potent cellular and humoral immunity. Accordingly, DCs are involved in the etiology of conditions as diverse as infectious diseases, allergic and autoimmune disorders, graft rejection and cancer. During the last decade, several methods have been developed to load DCs with tumor-associated antigens, ex vivo or in vivo, in the attempt to use them as therapeutic anticancer vaccines that would elicit clinically relevant immune responses. While this has not always been the case, several clinical studies have demonstrated that DC-based anticancer vaccines are capable of activating tumor-specific immune responses that increase overall survival, at least in a subset of patients. In 2010, this branch of clinical research has culminated with the approval by FDA of a DC-based therapeutic vaccine (sipuleucel-T, Provenge®) for use in patients with asymptomatic or minimally symptomatic metastatic hormone-refractory prostate cancer. Intense research efforts are currently dedicated to the identification of the immunological features of patients that best respond to DC-based anticancer vaccines. This knowledge may indeed lead to personalized combination strategies that would extend the benefit of DC-based immunotherapy to a larger patient population. In addition, widespread enthusiasm has been generated by the results of the first clinical trials based on in vivo DC targeting, an approach that holds great promises for the future of DC-based immunotherapy. In this Trial Watch, we will summarize the results of recently completed clinical trials and discuss the progress of ongoing studies that have evaluated/are evaluating DC-based interventions for cancer therapy.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France ; Institut Gustave Roussy; Villejuif, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Keunen O, Taxt T, Grüner R, Lund-Johansen M, Tonn JC, Pavlin T, Bjerkvig R, Niclou SP, Thorsen F. Multimodal imaging of gliomas in the context of evolving cellular and molecular therapies. Adv Drug Deliv Rev 2014; 76:98-115. [PMID: 25078721 DOI: 10.1016/j.addr.2014.07.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 07/14/2014] [Accepted: 07/22/2014] [Indexed: 01/18/2023]
Abstract
The vast majority of malignant gliomas relapse after surgery and standard radio-chemotherapy. Novel molecular and cellular therapies are thus being developed, targeting specific aspects of tumor growth. While histopathology remains the gold standard for tumor classification, neuroimaging has over the years taken a central role in the diagnosis and treatment follow up of brain tumors. It is used to detect and localize lesions, define the target area for biopsies, plan surgical and radiation interventions and assess tumor progression and treatment outcome. In recent years the application of novel drugs including anti-angiogenic agents that affect the tumor vasculature, has drastically modulated the outcome of brain tumor imaging. To properly evaluate the effects of emerging experimental therapies and successfully support treatment decisions, neuroimaging will have to evolve. Multi-modal imaging systems with existing and new contrast agents, molecular tracers, technological advances and advanced data analysis can all contribute to the establishment of disease relevant biomarkers that will improve disease management and patient care. In this review, we address the challenges of glioma imaging in the context of novel molecular and cellular therapies, and take a prospective look at emerging experimental and pre-clinical imaging techniques that bear the promise of meeting these challenges.
Collapse
|