101
|
Zhang X, Zhu S, Li T, Liu YJ, Chen W, Chen J. Targeting immune checkpoints in malignant glioma. Oncotarget 2017; 8:7157-7174. [PMID: 27756892 PMCID: PMC5351697 DOI: 10.18632/oncotarget.12702] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Accepted: 10/12/2016] [Indexed: 12/31/2022] Open
Abstract
Malignant glioma is the most common and a highly aggressive cancer in the central nervous system (CNS). Cancer immunotherapy, strategies to boost the body's anti-cancer immune responses instead of directly targeting tumor cells, recently achieved great success in treating several human solid tumors. Although once considered "immune privileged" and devoid of normal immunological functions, CNS is now considered a promising target for cancer immunotherapy, featuring the recent progresses in neurobiology and neuroimmunology and a highly immunosuppressive state in malignant glioma. In this review, we focus on immune checkpoint inhibitors, specifically, antagonizing monoclonal antibodies for programmed cell death protein-1 (PD-1), cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4), and indoleamine 2,3-dioxygenase (IDO). We discuss advances in the working mechanisms of these immune checkpoint molecules, their status in malignant glioma, and current preclinical and clinical trials targeting these molecules in malignant glioma.
Collapse
Affiliation(s)
- Xuhao Zhang
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Shan Zhu
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Tete Li
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Yong-Jun Liu
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
- Sanofi Research and Development, Cambridge, MA, USA
| | - Wei Chen
- ADC Biomedical Research Institute, Saint Paul, MN, USA
| | - Jingtao Chen
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| |
Collapse
|
102
|
Ohs I, van den Broek M, Nussbaum K, Münz C, Arnold SJ, Quezada SA, Tugues S, Becher B. Interleukin-12 bypasses common gamma-chain signalling in emergency natural killer cell lymphopoiesis. Nat Commun 2016; 7:13708. [PMID: 27982126 PMCID: PMC5172358 DOI: 10.1038/ncomms13708] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 10/27/2016] [Indexed: 12/26/2022] Open
Abstract
Differentiation and homeostasis of natural killer (NK) cells relies on common gamma-chain (γc)-dependent cytokines, in particular IL-15. Consequently, NK cells do not develop in mice with targeted γc deletion. Herein we identify an alternative pathway of NK-cell development driven by the proinflammatory cytokine IL-12, which can occur independently of γc-signalling. In response to viral infection or upon exogenous administration, IL-12 is sufficient to elicit the emergence of a population of CD122+CD49b+ cells by targeting NK-cell precursors (NKPs) in the bone marrow (BM). We confirm the NK-cell identity of these cells by transcriptome-wide analyses and their ability to eliminate tumour cells. Rather than using the conventional pathway of NK-cell development, IL-12-driven CD122+CD49b+ cells remain confined to a NK1.1lowNKp46low stage, but differentiate into NK1.1+NKp46+ cells in the presence of γc-cytokines. Our data reveal an IL-12-driven hard-wired pathway of emergency NK-cell lymphopoiesis bypassing steady-state γc-signalling.
Collapse
Affiliation(s)
- Isabel Ohs
- Inflammation Research, Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Maries van den Broek
- Tumor Immunology, Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Kathrin Nussbaum
- Inflammation Research, Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Sebastian J. Arnold
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, and BIOSS Centre of Biological Signalling Studies, Albert-Ludwigs-University, D-79104 Freiburg, Germany
- BIOSS Centre of Biological Signalling Studies, Albert-Ludwigs-University, D-79104 Freiburg, Germany
| | - Sergio A. Quezada
- Cancer Immunology Unit, Research Department of Hematology, University College London Cancer Institute, WC1E 6BT London, UK
| | - Sonia Tugues
- Inflammation Research, Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Burkhard Becher
- Inflammation Research, Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
103
|
Mirzaei R, Sarkar S, Yong VW. T Cell Exhaustion in Glioblastoma: Intricacies of Immune Checkpoints. Trends Immunol 2016; 38:104-115. [PMID: 27964820 DOI: 10.1016/j.it.2016.11.005] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/06/2016] [Accepted: 11/10/2016] [Indexed: 12/25/2022]
Abstract
Glioblastoma is an aggressive and incurable primary brain tumor. While the blockade of immune checkpoints leads to reversal of T cell exhaustion in many cancers, the efficacy of this therapy in glioblastoma requires further consideration of the brain microenvironment beyond T cell activity. Neural cells are crucially dependent on glucose for survival, and tumor cells rabidly consume glucose; the glucose-deprived microenvironment further elevates immune checkpoint molecules to benefit tumor growth and exacerbate T cell exhaustion. We review here how immune checkpoints drive exhaustion in T cells while favoring tumor metabolism, and discuss how glucose competition in the unique CNS milieu is an important consideration to improve the outcomes of immune checkpoint blockade in glioblastoma.
Collapse
Affiliation(s)
- Reza Mirzaei
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Susobhan Sarkar
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - V Wee Yong
- Department of Clinical Neurosciences and the Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
104
|
Kulig P, Musiol S, Freiberger SN, Schreiner B, Gyülveszi G, Russo G, Pantelyushin S, Kishihara K, Alessandrini F, Kündig T, Sallusto F, Hofbauer GFL, Haak S, Becher B. IL-12 protects from psoriasiform skin inflammation. Nat Commun 2016; 7:13466. [PMID: 27892456 PMCID: PMC5133729 DOI: 10.1038/ncomms13466] [Citation(s) in RCA: 142] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 10/03/2016] [Indexed: 02/08/2023] Open
Abstract
Neutralization of the common p40-subunit of IL-12/23 in psoriasis patients has led to a breakthrough in the management of moderate to severe disease. Aside from neutralizing IL-23, which is thought to be responsible for the curative effect, anti-p40 therapy also interferes with IL-12 signalling and type 1 immunity. Here we dissect the individual contribution of these two cytokines to the formation of psoriatic lesions and understand the effect of therapeutic co-targeting of IL-12 and IL-23 in psoriasis. Using a preclinical model for psoriatic plaque formation we show that IL-12, in contrast to IL-23, has a regulatory function by restraining the invasion of an IL-17-committed γδT (γδT17) cell subset. We discover that IL-12 receptor signalling in keratinocytes initiates a protective transcriptional programme that limits skin inflammation, suggesting that collateral targeting of IL-12 by anti-p40 monoclonal antibodies is counterproductive in the therapy of psoriasis.
Collapse
Affiliation(s)
- Paulina Kulig
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Stephanie Musiol
- Experimental Immunology Unit, Centre of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Centre Munich, 80802 Munich, Germany
| | | | - Bettina Schreiner
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Gabor Gyülveszi
- Institute for Research in Biomedicine, Cellular Immunology, 6500 Bellinzona, Switzerland
| | - Giancarlo Russo
- Functional Genomics Center Zurich, University of Zurich and ETH Zurich, 8057 Zurich, Switzerland
| | | | - Kenji Kishihara
- Department of Immunology, Faculty of Pharmaceutical Sciences, Nagasaki International University, 859-3298 Nagasaki, Japan
| | - Francesca Alessandrini
- Experimental Immunology Unit, Centre of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Centre Munich, 80802 Munich, Germany
| | - Thomas Kündig
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Federica Sallusto
- Institute for Research in Biomedicine, Cellular Immunology, 6500 Bellinzona, Switzerland
| | | | - Stefan Haak
- Experimental Immunology Unit, Centre of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Centre Munich, 80802 Munich, Germany
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
105
|
Smith SG, Baltz JL, Koppolu BP, Ravindranathan S, Nguyen K, Zaharoff DA. Immunological mechanisms of intravesical chitosan/interleukin-12 immunotherapy against murine bladder cancer. Oncoimmunology 2016; 6:e1259050. [PMID: 28197381 PMCID: PMC5283638 DOI: 10.1080/2162402x.2016.1259050] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 11/03/2016] [Accepted: 11/04/2016] [Indexed: 02/07/2023] Open
Abstract
There is a critical unmet clinical need for bladder cancer immunotherapies capable of inducing durable antitumor immunity. We have shown that four intravesical treatments with a simple co-formulation of interleukin-12 and the biopolymer chitosan not only destroy orthotopic bladder tumors, but also promote a potent long-lasting systemic immune response as evidenced through tumor-specific in vitro killing assays, complete protection from rechallenge, and abscopal antitumor responses at distant non-treated tumors. This study investigates the immunological kinetics underlying these results. We show through depletion studies that CD8+ T cells are required for initial tumor rejection, but CD4+ T cells protect against rechallenge. We also show that even a single intravesical treatment can eliminate tumors in 50% of mice with 6/9 and 7/8 mice eliminating tumors after three or four treatments respectively. We then performed immunophenotyping studies to analyze shifts in immune cell populations after each treatment within the tumor itself as well as in secondary lymphoid organs. These studies demonstrated an initial infiltration of macrophages and granulocytes followed by increased CD4+ and CD8+ effector-memory cells. This was coupled with a decreased level of regulatory T cells in peripheral lymph nodes as well as decreased myeloid-derived suppressor cell infiltration in the bladder. Taken together, these data demonstrate the ability of properly delivered interleukin-12-based therapies to engage adaptive immunity within the tumor itself as well as throughout the body and strengthen the case for clinical translation of chitosan/interleukin-12 as an intravesical treatment for bladder cancer.
Collapse
Affiliation(s)
- Sean G Smith
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, NC, USA; Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - John L Baltz
- Department of Biomedical Engineering, University of Arkansas , Fayetteville, AR, USA
| | - Bhanu Prasanth Koppolu
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, NC, USA; Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Sruthi Ravindranathan
- Department of Biomedical Engineering, University of Arkansas , Fayetteville, AR, USA
| | - Khue Nguyen
- Department of Cell and Molecular Biology, University of Arkansas , Fayetteville, AR, USA
| | - David A Zaharoff
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, NC, USA; Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| |
Collapse
|
106
|
Miska J, Rashidi A, Chang AL, Muroski ME, Han Y, Zhang L, Lesniak MS. Anti-GITR therapy promotes immunity against malignant glioma in a murine model. Cancer Immunol Immunother 2016; 65:1555-1567. [PMID: 27734112 DOI: 10.1007/s00262-016-1912-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/30/2016] [Indexed: 01/06/2023]
Abstract
Regulatory T cells (Tregs) are potently immunosuppressive cells that accumulate within the glioma microenvironment. The reduction in their function and/or trafficking has been previously shown to enhance survival in preclinical models of glioma. Glucocorticoid-induced TNFR-related protein (GITR) is a tumor necrosis factor superfamily receptor enriched on Tregs that has shown promise as a target for immunotherapy. An agonistic antibody against GITR has been demonstrated to inhibit Tregs in a number of models and has only been recently addressed in glioma. In this study, we examined the modality of the antibody function at the tumor site as opposed to the periphery as the blood-brain barrier prevents efficient antibody delivery to brain tumors. Mice harboring established GL261 tumors were treated with anti-GITR monotherapy and were shown to have a significant increase in overall survival (p < 0.01) when antibodies were injected directly into the glioma core, whereas peripheral antibody treatment only had a modest effect. Peripheral treatment resulted in a significant decrease in granzyme B (GrB) expression by Tregs, whereas intratumoral treatment resulted in both a decrease in GrB expression by Tregs and their selective depletion, which was largely mediated by FcγR-mediated destruction. We also discovered that anti-GITR treatment results in the enhanced survival and functionality of dendritic cells (DCs)-a previously unreported effect of this immunotherapy. In effect, this study demonstrates that the targeting of GITR is a feasible and noteworthy treatment option for glioma, but is largely dependent on the anatomical location in which the antibodies are delivered.
Collapse
Affiliation(s)
- Jason Miska
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Aida Rashidi
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Alan L Chang
- Committee on Cancer Biology, The University of Chicago, Chicago, IL, USA
| | - Megan E Muroski
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Yu Han
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Lingjiao Zhang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| | - Maciej S Lesniak
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N St. Clair, Suite 2210, Chicago, IL 60611, USA
| |
Collapse
|
107
|
Carter T, Shaw H, Cohn-Brown D, Chester K, Mulholland P. Ipilimumab and Bevacizumab in Glioblastoma. Clin Oncol (R Coll Radiol) 2016; 28:622-626. [PMID: 27169593 DOI: 10.1016/j.clon.2016.04.042] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 03/07/2016] [Accepted: 03/22/2016] [Indexed: 11/30/2022]
Abstract
The median survival in glioblastoma is just over a year, with no standard second-line therapy. Ipilimumab is an immune checkpoint inhibitor that activates the anti-tumour immune response by cytotoxic T-lymphocyte antigen-4 blockade. There is significant evidence supporting its role in the treatment of malignant melanoma, including in patients with brain metastases. The addition of the anti-angiogenesis agent, bevacizumab, seems to offer additional benefit and limit the immune-related side-effects of ipilimumab in melanoma. To date there have been no clinical trials investigating this combination in glioblastoma. In this single practice case series, 20 patients with glioblastoma were consented for and treated with ipilimumab and bevacizumab in combination. Safety, tolerability and the response to treatment were reviewed for all patients. Three patients were treated after palliative first-line radiotherapy, one patient after first-line chemoradiation and 16 patients were treated with recurrent disease. Sixty-five per cent of patients completed four cycles of 3 weekly ipilimumab therapy, administered with 2 weekly bevacizumab. Radiographic responses for patients with recurrent disease were evaluated by Response Assessment in Neuro-oncology (RANO) criteria; 31% of patients showed a partial response, 31% had stable disease and 38% had disease progression. The treatment combination was well tolerated, with treatment terminated before completion due to adverse events in two patients. Autoimmune toxicity was manageable with systemic corticosteroid therapy. Ipilimumab and bevacizumab in combination show promising activity with a predictable and manageable toxicity profile, warranting further clinical studies.
Collapse
Affiliation(s)
- T Carter
- UCL Cancer Institute, University College London, London, UK
| | - H Shaw
- University College London Hospital, London, UK
| | - D Cohn-Brown
- Harley Street at University College Hospital, London, UK
| | - K Chester
- UCL Cancer Institute, University College London, London, UK
| | - P Mulholland
- University College London Hospital, London, UK; Mount Vernon Cancer Centre, Northwood, Middlesex, UK.
| |
Collapse
|
108
|
Kamran N, Calinescu A, Candolfi M, Chandran M, Mineharu Y, Asad AS, Koschmann C, Nunez FJ, Lowenstein PR, Castro MG. Recent advances and future of immunotherapy for glioblastoma. Expert Opin Biol Ther 2016; 16:1245-64. [PMID: 27411023 PMCID: PMC5014608 DOI: 10.1080/14712598.2016.1212012] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 07/08/2016] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Outcome for glioma (GBM) remains dismal despite advances in therapeutic interventions including chemotherapy, radiotherapy and surgical resection. The overall survival benefit observed with immunotherapies in cancers such as melanoma and prostate cancer has fuelled research into evaluating immunotherapies for GBM. AREAS COVERED Preclinical studies have brought a wealth of information for improving the prognosis of GBM and multiple clinical studies are evaluating a wide array of immunotherapies for GBM patients. This review highlights advances in the development of immunotherapeutic approaches. We discuss the strategies and outcomes of active and passive immunotherapies for GBM including vaccination strategies, gene therapy, check point blockade and adoptive T cell therapies. We also focus on immunoediting and tumor neoantigens that can impact the efficacy of immunotherapies. EXPERT OPINION Encouraging results have been observed with immunotherapeutic strategies; some clinical trials are reaching phase III. Significant progress has been made in unraveling the molecular and genetic heterogeneity of GBM and its implications to disease prognosis. There is now consensus related to the critical need to incorporate tumor heterogeneity into the design of therapeutic approaches. Recent data also indicates that an efficacious treatment strategy will need to be combinatorial and personalized to the tumor genetic signature.
Collapse
Affiliation(s)
- Neha Kamran
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| | - Alexandra Calinescu
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| | - Marianela Candolfi
- c Instituto de Investigaciones Biomédicas (CONICET-UBA), Facultad de Medicina , Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Mayuri Chandran
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| | - Yohei Mineharu
- d Department of Neurosurgery , Kyoto University Graduate School of Medicine , Kyoto , Japan
| | - Antonela S Asad
- c Instituto de Investigaciones Biomédicas (CONICET-UBA), Facultad de Medicina , Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Carl Koschmann
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| | - Felipe J Nunez
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| | - Pedro R Lowenstein
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| | - Maria G Castro
- a Department of Neurosurgery , The University of Michigan School of Medicine , Ann Arbor , MI , USA
- b Department of Cell and Developmental Biology , The University of Michigan School of Medicine , Ann Arbor , MI , USA
| |
Collapse
|
109
|
Nduom EK, Weller M, Heimberger AB. Immunosuppressive mechanisms in glioblastoma. Neuro Oncol 2016; 17 Suppl 7:vii9-vii14. [PMID: 26516226 DOI: 10.1093/neuonc/nov151] [Citation(s) in RCA: 254] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Despite maximal surgical and medical therapy, the treatment of glioblastoma remains a seriously vexing problem, with median survival well under 2 years and few long-term survivors. Targeted therapy has yet to produce significant advances in treatment of these lesions in spite of advanced molecular characterization of glioblastoma and glioblastoma cancer stem cells. Recently, immunotherapy has emerged as a promising mode for some of the hardest to treat tumors, including metastatic melanoma. Although immunotherapy has been evaluated in glioblastoma in the past with limited success, better understanding of the failures of these therapies could lead to more successful treatments in the future. Furthermore, there is a persistent challenge for the use of immune therapy to treat glioblastoma secondary to the existence of redundant mechanisms of tumor-mediated immune suppression. Here we will address these mechanisms of immunosuppression in glioblastoma and therapeutic approaches.
Collapse
Affiliation(s)
- Edjah K Nduom
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (E.K.N., A.B.H.); Department of Neurology, University Hospital Zurich, Zurich, Switzerland (M.W.)
| | - Michael Weller
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (E.K.N., A.B.H.); Department of Neurology, University Hospital Zurich, Zurich, Switzerland (M.W.)
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas (E.K.N., A.B.H.); Department of Neurology, University Hospital Zurich, Zurich, Switzerland (M.W.)
| |
Collapse
|
110
|
Khan MN, Sharma AM, Pitz M, Loewen SK, Quon H, Poulin A, Essig M. High-grade glioma management and response assessment-recent advances and current challenges. ACTA ACUST UNITED AC 2016; 23:e383-91. [PMID: 27536188 DOI: 10.3747/co.23.3082] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The management of high-grade gliomas (hggs) is complex and ever-evolving. The standard of care for the treatment of hggs consists of surgery, chemotherapy, and radiotherapy. However, treatment options are influenced by multiple factors such as patient age and performance status, extent of tumour resection, biomarker profile, and tumour histology and grade. Follow-up cranial magnetic resonance imaging (mri) to differentiate treatment response from treatment effect can be challenging and affects clinical decision-making. An assortment of advanced radiologic techniques-including perfusion imaging with dynamic susceptibility contrast mri, dynamic contrast-enhanced mri, diffusion-weighted imaging, proton spectroscopy, mri subtraction imaging, and amino acid radiotracer imaging-can now incorporate novel physiologic data, providing new methods to help characterize tumour progression, pseudoprogression, and pseudoresponse. In the present review, we provide an overview of current treatment options for hgg and summarize recent advances and challenges in imaging technology.
Collapse
Affiliation(s)
- M N Khan
- Department of Radiology, University of Manitoba, Winnipeg, MB
| | - A M Sharma
- Department of Radiology, University of Manitoba, Winnipeg, MB;; Department of Radiation Oncology, CancerCare Manitoba, Winnipeg, MB
| | - M Pitz
- Department of Internal Medicine, University of Manitoba, Winnipeg, MB;; Department of Haematology and Medical Oncology, University of Manitoba, Winnipeg, MB
| | - S K Loewen
- Department of Radiology, University of Manitoba, Winnipeg, MB;; Department of Radiation Oncology, CancerCare Manitoba, Winnipeg, MB
| | - H Quon
- Department of Radiology, University of Manitoba, Winnipeg, MB;; Department of Radiation Oncology, CancerCare Manitoba, Winnipeg, MB
| | - A Poulin
- Department of Radiology, University of Manitoba, Winnipeg, MB;; Department of Radiology, Laval University, Quebec City, QC
| | - M Essig
- Department of Radiology, University of Manitoba, Winnipeg, MB
| |
Collapse
|
111
|
Neagu MR, Reardon DA. An Update on the Role of Immunotherapy and Vaccine Strategies for Primary Brain Tumors. Curr Treat Options Oncol 2016; 16:54. [PMID: 26454859 DOI: 10.1007/s11864-015-0371-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Existing therapies for glioblastoma (GBM), the most common malignant primary brain tumor in adults, have fallen short of improving the dismal patient outcomes, with an average 14-16-month median overall survival. The biological complexity and adaptability of GBM, redundancy of dysregulated signaling pathways, and poor penetration of therapies through the blood-brain barrier contribute to poor therapeutic progress. The current standard of care for newly diagnosed GBM consists of maximal safe resection, followed by fractionated radiotherapy combined with concurrent temozolomide (TMZ) and 6-12 cycles of adjuvant TMZ. At progression, bevacizumab with or without additional chemotherapy is an option for salvage therapy. The recent FDA approval of sipuleucel-T for prostate cancer and ipilumimab, nivolumab, and pembrolizumab for select solid tumors and the ongoing trials showing clinical efficacy and response durability herald a new era of cancer treatment with the potential to change standard-of-care treatment across multiple cancers. The evaluation of various immunotherapeutics is advancing for GBM, putting into question the dogma of the CNS as an immuno-privileged site. While the field is yet young, both active immunotherapy involving vaccine strategies and cellular therapy as well as reversal of GBM-induced global immune-suppression through immune checkpoint blockade are showing promising results and revealing essential immunological insights regarding kinetics of the immune response, immune evasion, and correlative biomarkers. The future holds exciting promise in establishing new treatment options for GBM that harness the patients' own immune system by activating it with immune checkpoint inhibitors, providing specificity using vaccine therapy, and allowing for modulation and enhancement by combinatorial approaches.
Collapse
Affiliation(s)
- Martha R Neagu
- Dana-Farber Cancer Institute, G4200, 44 Binney St, Boston, MA, 02115, USA
- Pappas Center for Neuro-Oncology, Massachusetts General Hospital, WACC 8-835m 55 Fruit St, Boston, MA, 02114, USA
| | - David A Reardon
- Dana-Farber Cancer Institute, G4200, 44 Binney St, Boston, MA, 02115, USA.
| |
Collapse
|
112
|
Lieberman NAP, Moyes KW, Crane CA. Developing immunotherapeutic strategies to target brain tumors. Expert Rev Anticancer Ther 2016; 16:775-88. [PMID: 27253692 DOI: 10.1080/14737140.2016.1192470] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Recent years have seen rapid growth in cancer treatments that enhance the anti-tumor activities of the immune system. Collectively known as immunotherapy, modulation of the immune system has shown success treating some hematological malignancies, but has yet to be successfully applied to the treatment of patients with brain tumors. AREAS COVERED This review highlights mechanistic insights from murine studies and compiled recent clinical trial data, focusing on the most aggressive brain tumor, glioblastoma (GBM). The field has recently accumulated a critical mass of data, and we discuss past treatment failures in the context of newly developed approaches now entering clinical trials. This article provides an overview of the immunotherapeutic armamentarium currently in development for the treatment of patients with GBM, who are in dire need of safe and effective therapies. Expert commentary: Themes that emerge include the importance of mitigating the effects of an immunosuppressive tumor microenvironment and the potential for innate immune cell activation to enhance cytotoxic anti-tumor activity. Consideration of these studies as a collective may inform the design of new immunotherapies, as well as the immune monitoring protocols for patients participating in clinical trials.
Collapse
Affiliation(s)
- Nicole A P Lieberman
- a Seattle Children's Research Institute, Ben Towne Center for Childhood Cancer Research , Seattle , WA , USA
| | - Kara White Moyes
- a Seattle Children's Research Institute, Ben Towne Center for Childhood Cancer Research , Seattle , WA , USA
| | - Courtney A Crane
- a Seattle Children's Research Institute, Ben Towne Center for Childhood Cancer Research , Seattle , WA , USA.,b Department of Neurological Surgery , University of Washington School of Medicine , Seattle , WA , USA
| |
Collapse
|
113
|
Mangani D, Weller M, Seyed Sadr E, Willscher E, Seystahl K, Reifenberger G, Tabatabai G, Binder H, Schneider H. Limited role for transforming growth factor-β pathway activation-mediated escape from VEGF inhibition in murine glioma models. Neuro Oncol 2016; 18:1610-1621. [PMID: 27286797 DOI: 10.1093/neuonc/now112] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 04/22/2016] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The vascular endothelial growth factor (VEGF) and transforming growth factor (TGF)-β pathways regulate key biological features of glioblastoma. Here we explore whether the TGF-β pathway, which promotes angiogenesis, invasiveness, and immunosuppression, acts as an escape pathway from VEGF inhibition. METHODS The role of the TGF-β pathway in escape from VEGF inhibition was assessed in vitro and in vivo and by gene expression profiling in syngeneic mouse glioma models. RESULTS We found that TGF-β is an upstream regulator of VEGF, whereas VEGF pathway activity does not alter the TGF-β pathway in vitro. In vivo, single-agent activity was observed for the VEGF antibody B20-4.1.1 in 3 and for the TGF-β receptor 1 antagonist LY2157299 in 2 of 4 models. Reduction of tumor volume and blood vessel density, but not induction of hypoxia, correlated with benefit from B20-4.1.1. Reduction of phosphorylated (p)SMAD2 by LY2157299 was seen in all models but did not predict survival. Resistance to B20 was associated with anti-angiogenesis escape pathway gene expression, whereas resistance to LY2157299 was associated with different immune response gene signatures in SMA-497 and GL-261 on transcriptomic profiling. The combination of B20 with LY2157299 was ineffective in SMA-497 but provided prolongation of survival in GL-261, associated with early suppression of pSMAD2 in tumor and host immune cells, prolonged suppression of angiogenesis, and delayed accumulation of tumor infiltrating microglia/macrophages. CONCLUSIONS Our study highlights the biological heterogeneity of murine glioma models and illustrates that cotargeting of the VEGF and TGF-β pathways might lead to improved tumor control only in subsets of glioblastoma.
Collapse
Affiliation(s)
- Davide Mangani
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (D.M., M.W., E.S.S., K.S., G.T., H.S.); Center for Neuroscience, University of Zurich, Zurich, Switzerland (M.W., G.T.); Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany (E.W., H.B.); Institute of Neuropathology, Heinrich Heine University, Düsseldorf, Germany (G.R.); German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Heidelberg, partner site, Essen/Düsseldorf, Germany (G.R.)
| | - Michael Weller
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (D.M., M.W., E.S.S., K.S., G.T., H.S.); Center for Neuroscience, University of Zurich, Zurich, Switzerland (M.W., G.T.); Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany (E.W., H.B.); Institute of Neuropathology, Heinrich Heine University, Düsseldorf, Germany (G.R.); German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Heidelberg, partner site, Essen/Düsseldorf, Germany (G.R.)
| | - Emad Seyed Sadr
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (D.M., M.W., E.S.S., K.S., G.T., H.S.); Center for Neuroscience, University of Zurich, Zurich, Switzerland (M.W., G.T.); Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany (E.W., H.B.); Institute of Neuropathology, Heinrich Heine University, Düsseldorf, Germany (G.R.); German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Heidelberg, partner site, Essen/Düsseldorf, Germany (G.R.)
| | - Edith Willscher
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (D.M., M.W., E.S.S., K.S., G.T., H.S.); Center for Neuroscience, University of Zurich, Zurich, Switzerland (M.W., G.T.); Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany (E.W., H.B.); Institute of Neuropathology, Heinrich Heine University, Düsseldorf, Germany (G.R.); German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Heidelberg, partner site, Essen/Düsseldorf, Germany (G.R.)
| | - Katharina Seystahl
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (D.M., M.W., E.S.S., K.S., G.T., H.S.); Center for Neuroscience, University of Zurich, Zurich, Switzerland (M.W., G.T.); Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany (E.W., H.B.); Institute of Neuropathology, Heinrich Heine University, Düsseldorf, Germany (G.R.); German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Heidelberg, partner site, Essen/Düsseldorf, Germany (G.R.)
| | - Guido Reifenberger
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (D.M., M.W., E.S.S., K.S., G.T., H.S.); Center for Neuroscience, University of Zurich, Zurich, Switzerland (M.W., G.T.); Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany (E.W., H.B.); Institute of Neuropathology, Heinrich Heine University, Düsseldorf, Germany (G.R.); German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Heidelberg, partner site, Essen/Düsseldorf, Germany (G.R.)
| | - Ghazaleh Tabatabai
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (D.M., M.W., E.S.S., K.S., G.T., H.S.); Center for Neuroscience, University of Zurich, Zurich, Switzerland (M.W., G.T.); Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany (E.W., H.B.); Institute of Neuropathology, Heinrich Heine University, Düsseldorf, Germany (G.R.); German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Heidelberg, partner site, Essen/Düsseldorf, Germany (G.R.)
| | - Hans Binder
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (D.M., M.W., E.S.S., K.S., G.T., H.S.); Center for Neuroscience, University of Zurich, Zurich, Switzerland (M.W., G.T.); Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany (E.W., H.B.); Institute of Neuropathology, Heinrich Heine University, Düsseldorf, Germany (G.R.); German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Heidelberg, partner site, Essen/Düsseldorf, Germany (G.R.)
| | - Hannah Schneider
- Laboratory of Molecular Neuro-Oncology, Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland (D.M., M.W., E.S.S., K.S., G.T., H.S.); Center for Neuroscience, University of Zurich, Zurich, Switzerland (M.W., G.T.); Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany (E.W., H.B.); Institute of Neuropathology, Heinrich Heine University, Düsseldorf, Germany (G.R.); German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Heidelberg, partner site, Essen/Düsseldorf, Germany (G.R.)
| |
Collapse
|
114
|
Lussier DM, Woolf EC, Johnson JL, Brooks KS, Blattman JN, Scheck AC. Enhanced immunity in a mouse model of malignant glioma is mediated by a therapeutic ketogenic diet. BMC Cancer 2016; 16:310. [PMID: 27178315 PMCID: PMC4866042 DOI: 10.1186/s12885-016-2337-7] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 05/04/2016] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Glioblastoma multiforme is a highly aggressive brain tumor with a poor prognosis, and advances in treatment have led to only marginal increases in overall survival. We and others have shown previously that the therapeutic ketogenic diet (KD) prolongs survival in mouse models of glioma, explained by both direct tumor growth inhibition and suppression of pro-inflammatory microenvironment conditions. The aim of this study is to assess the effects of the KD on the glioma reactive immune response. METHODS The GL261-Luc2 intracranial mouse model of glioma was used to investigate the effects of the KD on the tumor-specific immune response. Tumor-infiltrating CD8+ T cells, CD4+ T cells and natural killer (NK) cells were analyzed by flow cytometry. The expression of immune inhibitory receptors cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed death 1 (PD-1) on CD8+ T cells were also analyzed by flow cytometry. Analysis of intracellular cytokine production was used to determine production of IFN, IL-2 and IFN- in tumor-infiltrating CD8+ T and natural killer (NK) cells and IL-10 production by T regulatory cells. RESULTS We demonstrate that mice fed the KD had increased tumor-reactive innate and adaptive immune responses, including increased cytokine production and cytolysis via tumor-reactive CD8+ T cells. Additionally, we saw that mice maintained on the KD had increased CD4 infiltration, while T regulatory cell numbers stayed consistent. Lastly, mice fed the KD had a significant reduction in immune inhibitory receptor expression as well as decreased inhibitory ligand expression on glioma cells. CONCLUSIONS The KD may work in part as an immune adjuvant, boosting tumor-reactive immune responses in the microenvironment by alleviating immune suppression. This evidence suggests that the KD increases tumor-reactive immune responses, and may have implications in combinational treatment approaches.
Collapse
Affiliation(s)
- Danielle M Lussier
- School of Life Sciences, Arizona State University, Tempe, AZ, 85281, USA.,Center for Infectious Diseases and Vaccinology, Biodesign Institute, Arizona State University, Tempe, AZ, 85281, USA
| | - Eric C Woolf
- School of Life Sciences, Arizona State University, Tempe, AZ, 85281, USA.,Neuro-Oncology Research, Barrow Brain Tumor Research Center, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 W. Thomas Road, Phoenix, AZ, 85013, USA
| | - John L Johnson
- Center for Infectious Diseases and Vaccinology, Biodesign Institute, Arizona State University, Tempe, AZ, 85281, USA
| | - Kenneth S Brooks
- Neuro-Oncology Research, Barrow Brain Tumor Research Center, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 W. Thomas Road, Phoenix, AZ, 85013, USA
| | - Joseph N Blattman
- School of Life Sciences, Arizona State University, Tempe, AZ, 85281, USA.,Center for Infectious Diseases and Vaccinology, Biodesign Institute, Arizona State University, Tempe, AZ, 85281, USA
| | - Adrienne C Scheck
- School of Life Sciences, Arizona State University, Tempe, AZ, 85281, USA. .,Neuro-Oncology Research, Barrow Brain Tumor Research Center, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, 350 W. Thomas Road, Phoenix, AZ, 85013, USA.
| |
Collapse
|
115
|
Saito T, Nishikawa H, Wada H, Nagano Y, Sugiyama D, Atarashi K, Maeda Y, Hamaguchi M, Ohkura N, Sato E, Nagase H, Nishimura J, Yamamoto H, Takiguchi S, Tanoue T, Suda W, Morita H, Hattori M, Honda K, Mori M, Doki Y, Sakaguchi S. Two FOXP3(+)CD4(+) T cell subpopulations distinctly control the prognosis of colorectal cancers. Nat Med 2016; 22:679-84. [PMID: 27111280 DOI: 10.1038/nm.4086] [Citation(s) in RCA: 608] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 03/17/2016] [Indexed: 12/11/2022]
Abstract
CD4(+) T cells that express the forkhead box P3 (FOXP3) transcription factor function as regulatory T (Treg) cells and hinder effective immune responses against cancer cells. Abundant Treg cell infiltration into tumors is associated with poor clinical outcomes in various types of cancers. However, the role of Treg cells is controversial in colorectal cancers (CRCs), in which FOXP3(+) T cell infiltration indicated better prognosis in some studies. Here we show that CRCs, which are commonly infiltrated by suppression-competent FOXP3(hi) Treg cells, can be classified into two types by the degree of additional infiltration of FOXP3(lo) nonsuppressive T cells. The latter, which are distinguished from FOXP3(+) Treg cells by non-expression of the naive T cell marker CD45RA and instability of FOXP3, secreted inflammatory cytokines. Indeed, CRCs with abundant infiltration of FOXP3(lo) T cells showed significantly better prognosis than those with predominantly FOXP3(hi) Treg cell infiltration. Development of such inflammatory FOXP3(lo) non-Treg cells may depend on secretion of interleukin (IL)-12 and transforming growth factor (TGF)-β by tissues and their presence was correlated with tumor invasion by intestinal bacteria, especially Fusobacterium nucleatum. Thus, functionally distinct subpopulations of tumor-infiltrating FOXP3(+) T cells contribute in opposing ways to determining CRC prognosis. Depletion of FOXP3(hi) Treg cells from tumor tissues, which would augment antitumor immunity, could thus be used as an effective treatment strategy for CRCs and other cancers, whereas strategies that locally increase the population of FOXP3(lo) non-Treg cells could be used to suppress or prevent tumor formation.
Collapse
Affiliation(s)
- Takuro Saito
- Experimental Immunology, World Premier International Research Center (WPI), Immunology Frontier Research Center, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hiroyoshi Nishikawa
- Experimental Immunology, World Premier International Research Center (WPI), Immunology Frontier Research Center, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hisashi Wada
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yuji Nagano
- RIKEN Center for Integrative Medical Sciences (IMS), Kanagawa, Japan
| | - Daisuke Sugiyama
- Experimental Immunology, World Premier International Research Center (WPI), Immunology Frontier Research Center, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Koji Atarashi
- RIKEN Center for Integrative Medical Sciences (IMS), Kanagawa, Japan
| | - Yuka Maeda
- Experimental Immunology, World Premier International Research Center (WPI), Immunology Frontier Research Center, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Masahide Hamaguchi
- Experimental Immunology, World Premier International Research Center (WPI), Immunology Frontier Research Center, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Naganari Ohkura
- Experimental Immunology, World Premier International Research Center (WPI), Immunology Frontier Research Center, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Eiichi Sato
- Department of Pathology, Institute of Medical Science (Medical Research Center), Tokyo Medical University, Tokyo, Japan
| | - Hirotsugu Nagase
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Junichi Nishimura
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hirofumi Yamamoto
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shuji Takiguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Takeshi Tanoue
- RIKEN Center for Integrative Medical Sciences (IMS), Kanagawa, Japan
| | - Wataru Suda
- Graduate School of Frontier Sciences, University of Tokyo, Chiba, Japan
| | - Hidetoshi Morita
- School of Veterinary Medicine, Azabu University, Kanagawa, Japan
| | - Masahira Hattori
- Graduate School of Frontier Sciences, University of Tokyo, Chiba, Japan
| | - Kenya Honda
- RIKEN Center for Integrative Medical Sciences (IMS), Kanagawa, Japan
| | - Masaki Mori
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shimon Sakaguchi
- Experimental Immunology, World Premier International Research Center (WPI), Immunology Frontier Research Center, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
116
|
Hammerich L, Bhardwaj N, Kohrt HE, Brody JD. In situ vaccination for the treatment of cancer. Immunotherapy 2016; 8:315-30. [DOI: 10.2217/imt.15.120] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Vaccination has had a tremendous impact on human health by harnessing the immune system to prevent and eradicate infectious diseases and this same approach might be used in cancer therapy. Cancer vaccine development has been slowed hindered by the paucity of universal tumor-associated antigens and the difficulty in isolating and preparing individualized vaccines ex vivo. Another approach has been to initiate or stimulate an immune response in situ (at the tumor site) and thus exploit the potentially numerous tumor-associated antigens there. Here, we review the many approaches that have attempted to accomplish effective in situ vaccination, using intratumoral administration of immunomodulators to increase the numbers or activation state of either antigen present cells or T cells within the tumor.
Collapse
Affiliation(s)
- Linda Hammerich
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nina Bhardwaj
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Holbrook E Kohrt
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joshua D Brody
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
117
|
Hodges TR, Ferguson SD, Caruso HG, Kohanbash G, Zhou S, Cloughesy TF, Berger MS, Poste GH, Khasraw M, Ba S, Jiang T, Mikkelson T, Yung WKA, de Groot JF, Fine H, Cantley LC, Mellinghoff IK, Mitchell DA, Okada H, Heimberger AB. Prioritization schema for immunotherapy clinical trials in glioblastoma. Oncoimmunology 2016; 5:e1145332. [PMID: 27471611 DOI: 10.1080/2162402x.2016.1145332] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/12/2016] [Accepted: 01/16/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Emerging immunotherapeutic strategies for the treatment of glioblastoma (GBM) such as dendritic cell (DC) vaccines, heat shock proteins, peptide vaccines, and adoptive T-cell therapeutics, to name a few, have transitioned from the bench to clinical trials. With upcoming strategies and developing therapeutics, it is challenging to critically evaluate the practical, clinical potential of individual approaches and to advise patients on the most promising clinical trials. METHODS The authors propose a system to prioritize such therapies in an organized and data-driven fashion. This schema is based on four categories of factors: antigenic target robustness, immune-activation and -effector responses, preclinical vetting, and early evidence of clinical response. Each of these categories is subdivided to focus on the most salient elements for developing a successful immunotherapeutic approach for GBM, and a numerical score is generated. RESULTS The Score Card reveals therapeutics that have the most robust data to support their use, provides a reference prioritization score, and can be applied in a reiterative fashion with emerging data. CONCLUSIONS The authors hope that this schema will give physicians an evidence-based and rational framework to make the best referral decisions to better guide and serve this patient population.
Collapse
Affiliation(s)
- Tiffany R Hodges
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center , Houston, TX, USA
| | - Sherise D Ferguson
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center , Houston, TX, USA
| | - Hillary G Caruso
- The Division of Pediatrics, The University of Texas M.D. Anderson Cancer Center , Houston, TX, USA
| | - Gary Kohanbash
- Department of Neurosurgery, the University of California at San Francisco , San Francisco, USA
| | - Shouhao Zhou
- Department of Biostatistics, The University of Texas M.D. Anderson Cancer Center , Houston, TX, USA
| | - Timothy F Cloughesy
- Department of Neuro-Oncology, the University of California at Los Angeles , Los Angeles, CA, USA
| | - Mitchel S Berger
- Department of Neurosurgery, the University of California at San Francisco , San Francisco, USA
| | | | | | - Sujuan Ba
- The National Foundation for Cancer Research, Bethesda, MD, USA, Asian Fund for Cancer Research , Hong Kong, People's Republic of China
| | - Tao Jiang
- Department of Neurosurgery, Tiantan Hospital, Capital Medical University , Beijing, China
| | - Tom Mikkelson
- Department of Neurosurgery, Henry Ford Health System , Detroit, MI, USA
| | - W K Alfred Yung
- Department of Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center , Houston, TX, USA
| | - John F de Groot
- Department of Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center , Houston, TX, USA
| | - Howard Fine
- Division of Neuro-Oncology, Weill Cornell Medical College , New York, NY, USA
| | - Lewis C Cantley
- Department of Systems Biology, Harvard Medical School , Boston, MA, USA
| | - Ingo K Mellinghoff
- Department of Neurology and Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center , New York, NY, USA
| | - Duane A Mitchell
- Department of Neurosurgery, University of Florida , Gainesville, FL, USA
| | - Hideho Okada
- Department of Neurosurgery, the University of California at San Francisco , San Francisco, USA
| | - Amy B Heimberger
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center , Houston, TX, USA
| |
Collapse
|
118
|
Abstract
Gliomas are the most common primary brain tumors of the central nervous system, and carry a grim prognosis. Novel approaches utilizing the immune system as adjuvant therapy are quickly emerging as viable and effective options. Immunotherapeutic strategies being investigated to treat glioblastoma include: vaccination therapy targeted against either specific tumor antigens or whole tumor lysate, adoptive cellular therapy with cytotoxic T lymphocytes, chimeric antigen receptors and bi-specific T-cell engaging antibodies allowing circumvention of major histocompatibility complex restriction, aptamer therapy with aims for more efficient target delivery, and checkpoint blockade in order to release the tumor-mediated inhibition of the immune system. Given the heterogeneity of glioblastoma and its ability to gain mutations throughout the disease course, multifaceted treatment strategies utilizing multiple forms of immunotherapy in combination with conventional therapy will be most likely to succeed moving forward.
Collapse
Affiliation(s)
- Brandon D Liebelt
- Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Houston Methodist Neurological Institute, Houston, TX, USA
| | - Gaetano Finocchiaro
- Department of Neuro-oncology, IRCCS Istituto Neurologico Besta, Milan, Italy
| | - Amy B Heimberger
- Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
119
|
Ma X, Yan W, Zheng H, Du Q, Zhang L, Ban Y, Li N, Wei F. Regulation of IL-10 and IL-12 production and function in macrophages and dendritic cells. F1000Res 2015; 4. [PMID: 26918147 PMCID: PMC4754024 DOI: 10.12688/f1000research.7010.1] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/16/2015] [Indexed: 12/20/2022] Open
Abstract
Interleukin-10 and Interleukin-12 are produced primarily by pathogen-activated antigen-presenting cells, particularly macrophages and dendritic cells. IL-10 and IL-12 play very important immunoregulatory roles in host defense and immune homeostasis. Being anti- and pro-inflammatory in nature, respectively, their functions are antagonistically opposing. A comprehensive and in-depth understanding of their immunological properties and signaling mechanisms will help develop better clinical intervention strategies in therapy for a wide range of human disorders. Here, we provide an update on some emerging concepts, controversies, unanswered questions, and opinions regarding the immune signaling of IL-10 and IL-12.
Collapse
Affiliation(s)
- Xiaojing Ma
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, USA; Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY, USA
| | - Wenjun Yan
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, USA
| | - Hua Zheng
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, USA
| | - Qinglin Du
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, USA
| | - Lixing Zhang
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, USA
| | - Yi Ban
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY, USA
| | - Na Li
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, USA
| | - Fang Wei
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, USA
| |
Collapse
|
120
|
Hammerich L, Binder A, Brody JD. In situ vaccination: Cancer immunotherapy both personalized and off-the-shelf. Mol Oncol 2015; 9:1966-81. [PMID: 26632446 PMCID: PMC5528727 DOI: 10.1016/j.molonc.2015.10.016] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 10/20/2015] [Accepted: 10/20/2015] [Indexed: 01/15/2023] Open
Abstract
As cancer immunotherapy continues to benefit from novel approaches which cut immune 'brake pedals' (e.g. anti-PD1 and anti-CTLA4 antibodies) and push immune cell gas pedals (e.g. IL2, and IFNα) there will be increasing need to develop immune 'steering wheels' such as vaccines to guide the immune system specifically toward tumor associated antigens. Two primary hurdles in cancer vaccines have been: identification of universal antigens to be used in 'off-the-shelf' vaccines for common cancers, and 2) logistical hurdles of ex vivo production of individualized whole tumor cell vaccines. Here we summarize approaches using 'in situ vaccination' in which intratumoral administration of off-the-shelf immunomodulators have been developed to specifically induce (or amplify) T cell responses to each patient's individual tumor. Clinical studies have confirmed the induction of systemic immune and clinical responses to such approaches and preclinical models have suggested ways to further potentiate the translation of in situ vaccine trials for our patients.
Collapse
Affiliation(s)
- Linda Hammerich
- Icahn School of Medicine at Mount Sinai Hess Center for Science and Medicine, United States
| | - Adam Binder
- Icahn School of Medicine at Mount Sinai Hess Center for Science and Medicine, United States
| | - Joshua D Brody
- Icahn School of Medicine at Mount Sinai Hess Center for Science and Medicine, United States.
| |
Collapse
|
121
|
Kane JR, Miska J, Young JS, Kanojia D, Kim JW, Lesniak MS. Sui generis: gene therapy and delivery systems for the treatment of glioblastoma. Neuro Oncol 2015; 17 Suppl 2:ii24-ii36. [PMID: 25746089 DOI: 10.1093/neuonc/nou355] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Gene therapy offers a multidimensional set of approaches intended to treat and cure glioblastoma (GBM), in combination with the existing standard-of-care treatment (surgery and chemoradiotherapy), by capitalizing on the ability to deliver genes directly to the site of neoplasia to yield antitumoral effects. Four types of gene therapy are currently being investigated for their potential use in treating GBM: (i) suicide gene therapy, which induces the localized generation of cytotoxic compounds; (ii) immunomodulatory gene therapy, which induces or augments an enhanced antitumoral immune response; (iii) tumor-suppressor gene therapy, which induces apoptosis in cancer cells; and (iv) oncolytic virotherapy, which causes the lysis of tumor cells. The delivery of genes to the tumor site is made possible by means of viral and nonviral vectors for direct delivery of therapeutic gene(s), tumor-tropic cell carriers expressing therapeutic gene(s), and "intelligent" carriers designed to increase delivery, specificity, and tumoral toxicity against GBM. These vehicles are used to carry genetic material to the site of pathology, with the expectation that they can provide specific tropism to the desired site while limiting interaction with noncancerous tissue. Encouraging preclinical results using gene therapies for GBM have led to a series of human clinical trials. Although there is limited evidence of a therapeutic benefit to date, a number of clinical trials have convincingly established that different types of gene therapies delivered by various methods appear to be safe. Due to the flexibility of specialized carriers and genetic material, the technology for generating new and more effective therapies already exists.
Collapse
Affiliation(s)
- J Robert Kane
- Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Jason Miska
- Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Jacob S Young
- Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Deepak Kanojia
- Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Julius W Kim
- Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| | - Maciej S Lesniak
- Brain Tumor Center, The University of Chicago Pritzker School of Medicine, Chicago, Illinois
| |
Collapse
|
122
|
Reardon DA, Gokhale PC, Klein SR, Ligon KL, Rodig SJ, Ramkissoon SH, Jones KL, Conway AS, Liao X, Zhou J, Wen PY, Van Den Abbeele AD, Hodi FS, Qin L, Kohl NE, Sharpe AH, Dranoff G, Freeman GJ. Glioblastoma Eradication Following Immune Checkpoint Blockade in an Orthotopic, Immunocompetent Model. Cancer Immunol Res 2015; 4:124-35. [PMID: 26546453 DOI: 10.1158/2326-6066.cir-15-0151] [Citation(s) in RCA: 294] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 10/06/2015] [Indexed: 11/16/2022]
Abstract
Inhibition of immune checkpoints, including cytotoxic T-lymphocyte antigen-4 (CTLA-4), programmed death-1 (PD-1), and its ligand PD-L1, has demonstrated exciting and durable remissions across a spectrum of malignancies. Combinatorial regimens blocking complementary immune checkpoints further enhance the therapeutic benefit. The activity of these agents for patients with glioblastoma, a generally lethal primary brain tumor associated with significant systemic and microenvironmental immunosuppression, is not known. We therefore systematically evaluated the antitumor efficacy of murine antibodies targeting a broad panel of immune checkpoint molecules, including CTLA-4, PD-1, PD-L1, and PD-L2 when administered as single-agent therapy and in combinatorial regimens against an orthotopic, immunocompetent murine glioblastoma model. In these experiments, we observed long-term tumor-free survival following single-agent anti-PD-1, anti-PD-L1, or anti-CTLA-4 therapy in 50%, 20%, and 15% of treated animals, respectively. Combination therapy of anti-CTLA-4 plus anti-PD-1 cured 75% of the animals, even against advanced, later-stage tumors. In long-term survivors, tumor growth was not seen upon intracranial tumor rechallenge, suggesting that tumor-specific immune memory responses were generated. Inhibitory immune checkpoint blockade quantitatively increased activated CD8(+) and natural killer cells and decreased suppressive immune cells in the tumor microenvironment and draining cervical lymph nodes. Our results support prioritizing the clinical evaluation of PD-1, PD-L1, and CTLA-4 single-agent targeted therapy as well as combination therapy of CTLA-4 plus PD-1 blockade for patients with glioblastoma.
Collapse
Affiliation(s)
- David A Reardon
- Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts. Department of Medical Oncology, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts.
| | - Prafulla C Gokhale
- Belfer Institute for Applied Cancer Science, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts. Lurie Family Imaging Center, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts
| | - Sarah R Klein
- Department of Medical Oncology, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts
| | - Keith L Ligon
- Department of Medical Oncology, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts. Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts
| | - Scott J Rodig
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Shakti H Ramkissoon
- Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts
| | - Kristen L Jones
- Lurie Family Imaging Center, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts
| | - Amy Saur Conway
- Lurie Family Imaging Center, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts
| | - Xiaoyun Liao
- Department of Medical Oncology, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts
| | - Jun Zhou
- Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts
| | - Annick D Van Den Abbeele
- Lurie Family Imaging Center, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts. Department of Imaging, Dana-Farber Cancer Institute, Boston, Massachusetts. Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts
| | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts
| | - Lei Qin
- Lurie Family Imaging Center, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts
| | - Nancy E Kohl
- Lurie Family Imaging Center, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts
| | - Arlene H Sharpe
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts
| | - Glenn Dranoff
- Department of Medical Oncology, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
123
|
Dietrich PY, Dutoit V, Walker PR. Immunotherapy for glioma: from illusion to realistic prospects? Am Soc Clin Oncol Educ Book 2015:51-9. [PMID: 24857060 DOI: 10.14694/edbook_am.2014.34.51] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
There is now evidence that the rules established for tumor immunology and immunotherapy in general are relevant for brain tumors. Treatment strategies explored have mainly involved vaccines using either tumor cells or components, and vaccines with defined synthetic peptides. This latter approach offers the advantage to select well-characterized antigens with selective or preferential expression on glioma. This is a prerequisite because collateral damage to the brain is not allowed. A second strategy which is reaching clinical trials is T cell therapy using the patients' own lymphocytes engineered to become tumor reactive. Tumor specificity can be conferred by forced expression of either a high-avidity T cell receptor or an antitumor antibody (the latter cells are called chimeric antigen receptors). An advantage of T cell engineering is the possibility to modify the cells to augment cellular activation, in vivo persistence and resistance to the tumor immunosuppressive milieu. A direct targeting of the hostile glioma microenvironment will additionally be required for achieving potent immunotherapy and various trials are assessing this issue. Finally, combining immunotherapy with immune checkpoint inhibitors and chemotherapy must be explored within rigorous clinical trials that favor constant interactions between the bench and bedside. Regarding immunotherapy for glioma patients, what was an unrealistic dream a decade ago is today a credible prospect.
Collapse
Affiliation(s)
| | - Valérie Dutoit
- From the Center of Oncology, Geneva University Hospitals, Geneva, Switzerland
| | - Paul R Walker
- From the Center of Oncology, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
124
|
Śledzińska A, Menger L, Bergerhoff K, Peggs KS, Quezada SA. Negative immune checkpoints on T lymphocytes and their relevance to cancer immunotherapy. Mol Oncol 2015; 9:1936-65. [PMID: 26578451 DOI: 10.1016/j.molonc.2015.10.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 10/09/2015] [Accepted: 10/12/2015] [Indexed: 02/07/2023] Open
Abstract
The term 'inhibitory checkpoint' refers to the broad spectrum of co-receptors expressed by T cells that negatively regulate T cell activation thus playing a crucial role in maintaining peripheral self-tolerance. Co-inhibitory receptor ligands are highly expressed by a variety of malignancies allowing evasion of anti-tumour immunity. Recent studies demonstrate that manipulation of these co-inhibitory pathways can remove the immunological brakes that impede endogenous immune responses against tumours. Antibodies that block the interactions between co-inhibitory receptors and their ligands have delivered very promising clinical responses, as has been shown by recent successful trials targeting the CTLA-4 and PD-1 pathways. In this review, we discuss the mechanisms of action and expression pattern of co-inhibitory receptors on different T cells subsets, emphasising differences between CD4(+) and CD8(+) T cells. We also summarise recent clinical findings utilising immune checkpoint blockade.
Collapse
Affiliation(s)
- Anna Śledzińska
- Cancer Immunology Unit, UCL Cancer Institute, UCL, London, UK
| | - Laurie Menger
- Cancer Immunology Unit, UCL Cancer Institute, UCL, London, UK
| | | | - Karl S Peggs
- Cancer Immunology Unit, UCL Cancer Institute, UCL, London, UK.
| | | |
Collapse
|
125
|
O’Connor RA, Anderton SM. Inflammation-associated genes: risks and benefits to Foxp3+ regulatory T-cell function. Immunology 2015; 146:194-205. [PMID: 26190495 PMCID: PMC4582961 DOI: 10.1111/imm.12507] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 06/30/2015] [Accepted: 07/02/2015] [Indexed: 01/10/2023] Open
Abstract
Foxp3(+) regulatory T (Treg) cells prevent the development of autoimmunity and immunopathology, as well as maintaining homeostasis and tolerance to commensal microorganisms. The suppressive activity of Treg cells is their defining characteristic, generating great interest in their therapeutic potential. However, suppressive and effector functions are not entirely exclusive. Considerable evidence points to the ability of supposedly anti-inflammatory Foxp3-expressing Treg cells to also express transcription factors that have been characterized as cardinal drivers of T effector cell function. We will consider the mounting evidence that Treg cells can function in non-suppressive capacities and review the impetus for this functional change, its relevance to developing immune and autoimmune responses and its significance to the development of Treg-based therapies.
Collapse
Affiliation(s)
- Richard A O’Connor
- MRC Centre for Inflammation Research, Centre for Multiple Sclerosis Research and Centre for Immunity Infection and Evolution, University of EdinburghEdinburgh, UK
| | - Stephen M Anderton
- MRC Centre for Inflammation Research, Centre for Multiple Sclerosis Research and Centre for Immunity Infection and Evolution, University of EdinburghEdinburgh, UK
| |
Collapse
|
126
|
Chang MC, Chen YL, Chiang YC, Chen TC, Tang YC, Chen CA, Sun WZ, Cheng WF. Mesothelin-specific cell-based vaccine generates antigen-specific immunity and potent antitumor effects by combining with IL-12 immunomodulator. Gene Ther 2015; 23:38-49. [PMID: 26262583 DOI: 10.1038/gt.2015.85] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 06/12/2015] [Accepted: 06/23/2015] [Indexed: 12/30/2022]
Abstract
Ovarian cancer is a gynecologic malignancy with a high mortality rate. In the present study, we developed a novel cell-based vaccine, Meso-VAX, to generate mesothelin antigen-specific immune responses and immunotherapy against ovarian cancer. Mesothelin, a secreted protein anchored at the cell membrane, has recently been identified as a potential new tumor antigen for ovarian cancer. In this study, mice vaccinated with Meso-VAX and adeno-associated virus (AAV)-IL-12 exhibited dramatic increases in the number of mesothelin-specific CD4(+) helper and CD8(+) cytotoxic T-cell precursors, higher titers of anti-mesothelin Abs and in vitro tumor killing activity, and all of these mice were tumor-free after 60 days of tumor challenge. In addition, a significant reduction in peritoneal tumors and longer survival were noted in the mice vaccinated with Meso-VAX combined with AAV-IL-12. CD4(+) helper and CD8(+) cytotoxic T lymphocytes were essential for the antitumor effect generated by Meso-VAX combined with AAV-IL-12. The post-vaccination sera of the mice vaccinated with Meso-VAX and AAV-IL-12 also showed mesothelin-specific complement-dependent cell-mediated cytotoxicity. Our results suggest that a Meso-VAX cell-based vaccine combined with AAV-IL-12 can generate antigen-specific immunological responses and antitumor effects on ovarian cancer.
Collapse
Affiliation(s)
- M-C Chang
- Department of Obstetrics and Gynecology, Medicine College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Anesthesiology, Medicine College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Y-L Chen
- Department of Obstetrics and Gynecology, Cathay General Hospital, Taipei, Taiwan
| | - Y-C Chiang
- Department of Obstetrics and Gynecology, Medicine College of Medicine, National Taiwan University, Taipei, Taiwan
| | - T-C Chen
- Department of Obstetrics and Gynecology, Medicine College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Y-C Tang
- Department of Obstetrics and Gynecology, Medicine College of Medicine, National Taiwan University, Taipei, Taiwan
| | - C-A Chen
- Department of Obstetrics and Gynecology, Medicine College of Medicine, National Taiwan University, Taipei, Taiwan
| | - W-Z Sun
- Department of Anesthesiology, Medicine College of Medicine, National Taiwan University, Taipei, Taiwan
| | - W-F Cheng
- Department of Obstetrics and Gynecology, Medicine College of Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, Medicine College of Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Oncology, Medicine College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
127
|
Abdi K, Singh NJ. Making many from few: IL-12p40 as a model for the combinatorial assembly of heterodimeric cytokines. Cytokine 2015; 76:53-7. [PMID: 26242928 DOI: 10.1016/j.cyto.2015.07.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 07/28/2015] [Accepted: 07/29/2015] [Indexed: 12/20/2022]
Abstract
How dendritic cells (DCs) gather information from the local milieu at a site of infection or injury and communicate this to influence adaptive immunity is not well understood. We and others have reported that soon after microbial encounter, DCs secrete the p40 subunit of IL-12, by itself, in a monomeric form. Based on recent data that this p40 monomer subsequently associates with p35 released from other cells to generate functional IL-12, we proposed that p40 can function as a DC-derived probe which samples the composition of the local milieu by looking for other binding partners. In this opinion, we discuss how such a sampling function might generate an elaborate combinatorial "code" of heterodimeric cytokines, capable of conveying location-specific information to cells downstream of DC activation, including NK and T cells.
Collapse
Affiliation(s)
- Kaveh Abdi
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Twinbrook II 12441 Parklawn Dr., Rockville, MD 20852, United States
| | - Nevil J Singh
- Department of Microbiology & Immunology, University of Maryland School of Medicine, 685 W Baltimore St, HSF1, Room 380, Baltimore, MD 21201, United States.
| |
Collapse
|
128
|
Bransi A, Salgado OC, Beffinger M, Milo K, Silina K, Yagita H, Becher B, Knuth A, van den Broek M. Rational Combination of Immunotherapies with Clinical Efficacy in Mice with Advanced Cancer. Cancer Immunol Res 2015; 3:1279-88. [PMID: 26141620 DOI: 10.1158/2326-6066.cir-15-0103-t] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 07/01/2015] [Indexed: 11/16/2022]
Abstract
In the context of cancer, naïve T cells are insufficiently primed and become progressively dysfunctional. Boosting antitumor responses by blocking PD-1 or CTLA-4 results in durable clinical responses only in a limited proportion of cancer patients, suggesting that other pathways must be targeted to improve clinical efficacy. Our preclinical study in TRAMP mice comparing 14 different immune interventions identified anti-CD40 + IL2/anti-IL2 complexes + IL12Fc as a uniquely efficacious treatment that prevents tolerance induction, promotes priming of sustained, protective tumor-specific CD8(+) T cells, and cures late-stage cancer when given together with adoptively transferred tumor-specific T cells. We propose that improving signals 2 (costimulation) and 3 (cytokines) together with fresh tumor-specific, rather than boosting of dysfunctional preexisting memory, T cells represents a potent therapy for advanced cancer.
Collapse
Affiliation(s)
- Ali Bransi
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | | - Michal Beffinger
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Karim Milo
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Karina Silina
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Alexander Knuth
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Maries van den Broek
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
129
|
Zundler S, Neurath MF. Interleukin-12: Functional activities and implications for disease. Cytokine Growth Factor Rev 2015; 26:559-68. [PMID: 26182974 DOI: 10.1016/j.cytogfr.2015.07.003] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 07/01/2015] [Indexed: 02/01/2023]
Abstract
Interleukin-12 (IL-12) was the first member of the IL-12 family of cytokines to be identified and has therefore become its eponym. It is a heterodimeric protein of two subunits (p35, p40) secreted by phagocytic cells in response to pathogens and mainly acts through STAT4 to induce IFN-γ production in T and NK cells. IFN-γ in turn mediates proinflammatory functions and activates T-bet. As IL-12 engages in TH1 development, it is believed to represent an important link between innate and adaptive immunity. Following its identification and the finding of its association to TH1 commitment, great hopes were placed in IL-12 to become a target for therapeutic applications in multiple settings of autoimmunity and cancer. Though, the discovery of the related members of the IL-12 family and several rather disappointing attempts to translate experimental results into clinical practice, have relativized these hopes. Nevertheless, IL-12 remains a cytokine of outstanding importance with lots of unresolved questions. In this review, we will first briefly depict the biochemistry of the cytokine, its receptor and the related signal transduction, before summarizing the regulation of IL-12 production and its biological functions. We will then describe the current knowledge about the implication of IL-12 in different murine disease models as well as in the corresponding human conditions and comment on possible consequences for future clinical applications.
Collapse
Affiliation(s)
- Sebastian Zundler
- Department of Medicine 1, University of Erlangen-Nuremberg, Kussmaul Campus for Medical Research & Translational Research Center, Medical Clinic 1, Ulmenweg 18, 91054 Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, University of Erlangen-Nuremberg, Kussmaul Campus for Medical Research & Translational Research Center, Medical Clinic 1, Ulmenweg 18, 91054 Erlangen, Germany.
| |
Collapse
|
130
|
Börschel N, Schwöppe C, Zerbst C, Angenendt L, Kessler T, Klapper W, Giovannoni L, Elia G, Neri D, Berdel WE, Mesters RM, Schliemann C. Potentiating the activity of rituximab against mantle cell lymphoma in mice by targeting interleukin-2 to the neovasculature. Leuk Res 2015; 39:739-48. [DOI: 10.1016/j.leukres.2015.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 03/10/2015] [Accepted: 04/03/2015] [Indexed: 11/28/2022]
|
131
|
Singh M, Overwijk WW. Intratumoral immunotherapy for melanoma. Cancer Immunol Immunother 2015; 64:911-21. [PMID: 26050024 PMCID: PMC11028428 DOI: 10.1007/s00262-015-1727-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 05/29/2015] [Indexed: 12/27/2022]
Abstract
Selection of suitable tumor-associated antigens is a major challenge in the development of effective cancer vaccines. Intratumoral (i.t.) immunotherapy empowers the immune system to mount T cell responses against tumor-associated antigens which are most immunogenic. To mediate systemic tumor regression, i.t. immunotherapy must generate systemic T cell responses that can target distant metastases beyond the initially treated tumor mass. Now that promising preclinical results and some initial success in clinical trials have been obtained, we here review i.t. immunotherapy-related preclinical and clinical studies, their mechanisms of action and future prospects.
Collapse
Affiliation(s)
- Manisha Singh
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 7455 Fannin St., Unit 0904, Houston, TX 77030 USA
| | - Willem W. Overwijk
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 7455 Fannin St., Unit 0904, Houston, TX 77030 USA
- The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX USA
| |
Collapse
|
132
|
Teng MWL, Bowman EP, McElwee JJ, Smyth MJ, Casanova JL, Cooper AM, Cua DJ. IL-12 and IL-23 cytokines: from discovery to targeted therapies for immune-mediated inflammatory diseases. Nat Med 2015; 21:719-29. [PMID: 26121196 DOI: 10.1038/nm.3895] [Citation(s) in RCA: 572] [Impact Index Per Article: 63.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 05/05/2015] [Indexed: 12/18/2022]
Abstract
The cytokine interleukin-12 (IL-12) was thought to have a central role in T cell-mediated responses in inflammation for more than a decade after it was first identified. Discovery of the cytokine IL-23, which shares a common p40 subunit with IL-12, prompted efforts to clarify the relative contribution of these two cytokines in immune regulation. Ustekinumab, a therapeutic agent targeting both cytokines, was recently approved to treat psoriasis and psoriatic arthritis, and related agents are in clinical testing for a variety of inflammatory disorders. Here we discuss the therapeutic rationale for targeting these cytokines, the unintended consequences for host defense and tumor surveillance and potential ways in which these therapies can be applied to treat additional immune disorders.
Collapse
Affiliation(s)
- Michele W L Teng
- 1] Cancer Immunoregulation and Immunotherapy and Immunology in Cancer and Infection Laboratories, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia. [2] School of Medicine, University of Queensland, Herston, Queensland, Australia
| | | | | | - Mark J Smyth
- 1] Cancer Immunoregulation and Immunotherapy and Immunology in Cancer and Infection Laboratories, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia. [2] School of Medicine, University of Queensland, Herston, Queensland, Australia
| | - Jean-Laurent Casanova
- 1] St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA. [2] Howard Hughes Medical Institute, New York, New York, USA. [3] Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, Paris, France. [4] Pediatric Hematology and Immunology Unit, Necker Hospital for Sick Children, Paris, France. [5] Paris Descartes University, Imagine Institute, Paris, France
| | | | - Daniel J Cua
- Merck Research Laboratories, Palo Alto, California, USA
| |
Collapse
|
133
|
Klinke DJ. Enhancing the discovery and development of immunotherapies for cancer using quantitative and systems pharmacology: Interleukin-12 as a case study. J Immunother Cancer 2015; 3:27. [PMID: 26082838 PMCID: PMC4468964 DOI: 10.1186/s40425-015-0069-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 04/28/2015] [Indexed: 12/22/2022] Open
Abstract
Recent clinical successes of immune checkpoint modulators have unleashed a wave of enthusiasm associated with cancer immunotherapy. However, this enthusiasm is dampened by persistent translational hurdles associated with cancer immunotherapy that mirror the broader pharmaceutical industry. Specifically, the challenges associated with drug discovery and development stem from an incomplete understanding of the biological mechanisms in humans that are targeted by a potential drug and the financial implications of clinical failures. Sustaining progress in expanding the clinical benefit provided by cancer immunotherapy requires reliably identifying new mechanisms of action. Along these lines, quantitative and systems pharmacology (QSP) has been proposed as a means to invigorate the drug discovery and development process. In this review, I discuss two central themes of QSP as applied in the context of cancer immunotherapy. The first theme focuses on a network-centric view of biology as a contrast to a "one-gene, one-receptor, one-mechanism" paradigm prevalent in contemporary drug discovery and development. This theme has been enabled by the advances in wet-lab capabilities to assay biological systems at increasing breadth and resolution. The second theme focuses on integrating mechanistic modeling and simulation with quantitative wet-lab studies. Drawing from recent QSP examples, large-scale mechanistic models that integrate phenotypic signaling-, cellular-, and tissue-level behaviors have the potential to lower many of the translational hurdles associated with cancer immunotherapy. These include prioritizing immunotherapies, developing mechanistic biomarkers that stratify patient populations and that reflect the underlying strength and dynamics of a protective host immune response, and facilitate explicit sharing of our understanding of the underlying biology using mechanistic models as vehicles for dialogue. However, creating such models require a modular approach that assumes that the biological networks remain similar in health and disease. As oncogenesis is associated with re-wiring of these biological networks, I also describe an approach that combines mechanistic modeling with quantitative wet-lab experiments to identify ways in which malignant cells alter these networks, using Interleukin-12 as an example. Collectively, QSP represents a new holistic approach that may have profound implications for how translational science is performed.
Collapse
Affiliation(s)
- David J Klinke
- Department of Chemical Engineering and Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 25606 USA
| |
Collapse
|
134
|
Abstract
Immunotherapy is coming to the fore as a viable anti-cancer treatment modality, even in poorly immunogenic cancers such as glioblastoma (GBM). Accumulating evidence suggests that the central nervous system may not be impervious to tumor-specific immune cells and could be an adequate substrate for immunologic anti-cancer therapies. Recent advances in antigen-specific cancer vaccines and checkpoint blockade in GBM provide promise for future immunotherapy in glioma. As anti-GBM immunotherapeutics enter clinical trials, it is important to understand the interactions, if any, between immune-based treatment modalities and the current standard of care for GBM involving chemoradiation and steroid therapy. Current data suggests that chemoradiation may not preclude the success of immunotherapeutics, as their effects may be synergistic. The future of therapy for GBM lies in the power of combination modalities, involving immunotherapy and the current standard of care.
Collapse
|
135
|
Yan J, Kong LY, Hu J, Gabrusiewicz K, Dibra D, Xia X, Heimberger AB, Li S. FGL2 as a Multimodality Regulator of Tumor-Mediated Immune Suppression and Therapeutic Target in Gliomas. J Natl Cancer Inst 2015; 107:djv137. [PMID: 25971300 DOI: 10.1093/jnci/djv137] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Fibrinogen-like protein 2 (FGL2) may promote glioblastoma multiforme (GBM) cancer development by inducing multiple immune-suppression mechanisms. METHODS The biological significance of FGL2 expression was assessed using the The Cancer Genome Atlast (TCGA) glioma database and tumor lysates analysis. The therapeutic effects of an anti-Fgl2 antibody and the role of immune suppression regulation by Fgl2 were determined in immune-competent, NOD-scid IL2Rgammanull (NSG), and FcɣRIIB-/- mice (n = 3-18 per group). Data were analyzed with two-way analysis of variance, log-rank survival analysis, and Pearson correlation. All statistical tests were two-sided. RESULTS In low-grade gliomas, 72.5% of patients maintained two copies of the FGL2 gene, whereas 83.8% of GBM patients had gene amplification or copy gain. Patients with high levels of FGL2 mRNA in glioma tissues had a lower overall survival (P = .009). Protein levels of FGL2 in GBM lysates were higher relative to low-grade glioma lysates (11.48±5.75ng/mg vs 3.96±1.01ng/mg, P = .003). In GL261 mice treated with an anti-FGL2 antibody, median survival was 27 days compared with only 17 days for mice treated with an isotype control antibody (P = .01). The anti-FGL2 antibody treatment reduced CD39(+) Tregs, M2 macrophages, programmed cell death protein 1 (PD-1), and myeloid-derived suppressor cells (MDSCs). FGL2-induced increases in M2, CD39, and PD-1 were ablated in FcɣRIIB-/- mice. CONCLUSIONS FGL2 augments glioma immunosuppression by increasing the expression levels of PD-1 and CD39, expanding the frequency of tumor-supportive M2 macrophages via the FcγRIIB pathway, and enhancing the number of MDSCs and CD39(+) regulatory T cells. Collectively, these results show that FGL2 functions as a key immune-suppressive modulator and has potential as an immunotherapeutic target for treating GBM.
Collapse
Affiliation(s)
- Jun Yan
- Department of Pediatric Research (JY, JH, DD, XX, SL) and Department of Neurosurgery (LYK, KG, ABH), The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Ling-Yuan Kong
- Department of Pediatric Research (JY, JH, DD, XX, SL) and Department of Neurosurgery (LYK, KG, ABH), The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Jiemiao Hu
- Department of Pediatric Research (JY, JH, DD, XX, SL) and Department of Neurosurgery (LYK, KG, ABH), The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Konrad Gabrusiewicz
- Department of Pediatric Research (JY, JH, DD, XX, SL) and Department of Neurosurgery (LYK, KG, ABH), The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Denada Dibra
- Department of Pediatric Research (JY, JH, DD, XX, SL) and Department of Neurosurgery (LYK, KG, ABH), The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Xueqing Xia
- Department of Pediatric Research (JY, JH, DD, XX, SL) and Department of Neurosurgery (LYK, KG, ABH), The University of Texas M.D. Anderson Cancer Center, Houston, TX
| | - Amy B Heimberger
- Department of Pediatric Research (JY, JH, DD, XX, SL) and Department of Neurosurgery (LYK, KG, ABH), The University of Texas M.D. Anderson Cancer Center, Houston, TX.
| | - Shulin Li
- Department of Pediatric Research (JY, JH, DD, XX, SL) and Department of Neurosurgery (LYK, KG, ABH), The University of Texas M.D. Anderson Cancer Center, Houston, TX.
| |
Collapse
|
136
|
Bausch-Fluck D, Hofmann A, Bock T, Frei AP, Cerciello F, Jacobs A, Moest H, Omasits U, Gundry RL, Yoon C, Schiess R, Schmidt A, Mirkowska P, Härtlová A, Van Eyk JE, Bourquin JP, Aebersold R, Boheler KR, Zandstra P, Wollscheid B. A mass spectrometric-derived cell surface protein atlas. PLoS One 2015; 10:e0121314. [PMID: 25894527 PMCID: PMC4404347 DOI: 10.1371/journal.pone.0121314] [Citation(s) in RCA: 284] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 01/30/2015] [Indexed: 01/08/2023] Open
Abstract
Cell surface proteins are major targets of biomedical research due to their utility as cellular markers and their extracellular accessibility for pharmacological intervention. However, information about the cell surface protein repertoire (the surfaceome) of individual cells is only sparsely available. Here, we applied the Cell Surface Capture (CSC) technology to 41 human and 31 mouse cell types to generate a mass-spectrometry derived Cell Surface Protein Atlas (CSPA) providing cellular surfaceome snapshots at high resolution. The CSPA is presented in form of an easy-to-navigate interactive database, a downloadable data matrix and with tools for targeted surfaceome rediscovery (http://wlab.ethz.ch/cspa). The cellular surfaceome snapshots of different cell types, including cancer cells, resulted in a combined dataset of 1492 human and 1296 mouse cell surface glycoproteins, providing experimental evidence for their cell surface expression on different cell types, including 136 G-protein coupled receptors and 75 membrane receptor tyrosine-protein kinases. Integrated analysis of the CSPA reveals that the concerted biological function of individual cell types is mainly guided by quantitative rather than qualitative surfaceome differences. The CSPA will be useful for the evaluation of drug targets, for the improved classification of cell types and for a better understanding of the surfaceome and its concerted biological functions in complex signaling microenvironments.
Collapse
Affiliation(s)
- Damaris Bausch-Fluck
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
- Department of Health Sciences and Technology, BMPP, ETH Zurich, Zurich, Switzerland
| | - Andreas Hofmann
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Thomas Bock
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Andreas P. Frei
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Ferdinando Cerciello
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
- Laboratory of Molecular Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Andrea Jacobs
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Hansjoerg Moest
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Ulrich Omasits
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
- Department of Health Sciences and Technology, BMPP, ETH Zurich, Zurich, Switzerland
| | - Rebekah L. Gundry
- Department of Biochemistry, Medical College of Wisconsin, Wisconsin, Milwaukee, United States of America
| | - Charles Yoon
- Institute for Biomaterials & Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Ralph Schiess
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Alexander Schmidt
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Paulina Mirkowska
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
- Oncology Research Laboratory, University Children Hospital Zurich, Zurich, Switzerland
| | - Anetta Härtlová
- Centre of Advanced Studies, Faculty of Military Health Sciences, University of Defense, Hradec Kralove, Czech Republic
| | - Jennifer E. Van Eyk
- Department of Medicine, Biological Chemistry and Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jean-Pierre Bourquin
- Oncology Research Laboratory, University Children Hospital Zurich, Zurich, Switzerland
| | - Ruedi Aebersold
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
- Center for Systems Physiology and Metabolic Diseases, Zurich, Switzerland
- Faculty of Science, University of Zurich, Zurich, Switzerland
| | - Kenneth R. Boheler
- SCRMC, LKS Faculty of Medicine, Hong Kong University, Hong Kong, Hong Kong SAR
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Peter Zandstra
- Institute for Biomaterials & Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Bernd Wollscheid
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
- Department of Health Sciences and Technology, BMPP, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
137
|
Gu J, Zhang L, Wang Z, Chen Y, Zhang G, Zhang D, Wang X, Bai X, Li X, Lili Z. The Effect of Asarinin on Toll-Like Pathway in Rats After Cardiac Allograft Implantation. Transplant Proc 2015; 47:545-8. [DOI: 10.1016/j.transproceed.2014.12.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Revised: 12/02/2014] [Accepted: 12/31/2014] [Indexed: 10/23/2022]
|
138
|
Biomarkers for glioma immunotherapy: the next generation. J Neurooncol 2015; 123:359-72. [PMID: 25724916 DOI: 10.1007/s11060-015-1746-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 02/16/2015] [Indexed: 12/11/2022]
Abstract
The term "biomarker" historically refers to a single parameter, such as the expression level of a gene or a radiographic pattern, used to indicate a broader biological state. Molecular indicators have been applied to several aspects of cancer therapy: to describe the genotypic and phenotypic state of neoplastic tissue for prognosis, to predict susceptibility to anti-proliferative agents, to validate the presence of specific drug targets, and to evaluate responsiveness to therapy. For glioblastoma (GBM), immunohistochemical and radiographic biomarkers accessible to the clinical lab have informed traditional regimens, but while immunotherapies have emerged as potentially disruptive weapons against this diffusely infiltrating, heterogeneous tumor, biomarkers with strong predictive power have not been fully established. The cancer immunotherapy field, through the recently accelerated expansion of trials, is currently leveraging this wealth of clinical and biological data to define and revise the use of biomarkers for improving prognostic accuracy, personalization of therapy, and evaluation of responses across the wide variety of tumors. Technological advancements in DNA sequencing, cytometry, and microscopy have facilitated the exploration of more integrated, high-dimensional profiling of the disease system-incorporating both immune and tumor parameters-rather than single metrics, as biomarkers for therapeutic sensitivity. Here we discuss the utility of traditional GBM biomarkers in immunotherapy and how the impending transformation of the biomarker paradigm-from single markers to integrated profiles-may offer the key to bringing predictive, personalized immunotherapy to GBM patients.
Collapse
|
139
|
The interplay of effector and regulatory T cells in cancer. Curr Opin Immunol 2015; 33:101-11. [PMID: 25728990 DOI: 10.1016/j.coi.2015.02.003] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 01/19/2015] [Accepted: 02/06/2015] [Indexed: 01/05/2023]
Abstract
Regulatory T (Treg) cells suppress effector T (Teff) cells and prevent immune-mediated rejection of cancer. Much less appreciated are mechanisms by which Teff cells antagonize Treg cells. Herein, we consider how complex reciprocal interactions between Teff and Treg cells shape their population dynamics within tumors. Under states of tolerance, including during tumor escape, suppressed Teff cells support Treg cell populations through antigen-dependent provision of interleukin (IL)-2. During immune activation, Teff cells can lose this supportive capacity and directly antagonize Treg cell populations to neutralize their immunosuppressive function. While this latter state is rarely achieved spontaneously within tumors, we propose that therapeutic induction of immune activation has the potential to stably disrupt immunosuppressive population states resulting in durable cancer regression.
Collapse
|
140
|
Han J, Alvarez-Breckenridge CA, Wang QE, Yu J. TGF-β signaling and its targeting for glioma treatment. Am J Cancer Res 2015; 5:945-955. [PMID: 26045979 PMCID: PMC4449428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Accepted: 02/19/2015] [Indexed: 06/04/2023] Open
Abstract
Transforming growth factor-beta (TGF-β) is a pleiotropic cytokine, secreted by a variety of cells including immune cells, tumor cells, and stromal cells. TGF-β signaling is dysregulated in cancer patients, and this aberrant signaling at least in part contributes to initiation and progression of many cancers including glioma. The dysregulated signaling components provide molecular targets for the treatment of glioma. In this article, we review TGF-β signaling and its targeting in glioma.
Collapse
Affiliation(s)
- Jianfeng Han
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State UniversityColumbus, Ohio 43210, USA
- The Ohio State University Comprehensive Cancer CenterColumbus, Ohio 43210, USA
| | | | - Qi-En Wang
- The Ohio State University Comprehensive Cancer CenterColumbus, Ohio 43210, USA
| | - Jianhua Yu
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State UniversityColumbus, Ohio 43210, USA
- The Ohio State University Comprehensive Cancer CenterColumbus, Ohio 43210, USA
| |
Collapse
|
141
|
Van der Jeught K, Bialkowski L, Daszkiewicz L, Broos K, Goyvaerts C, Renmans D, Van Lint S, Heirman C, Thielemans K, Breckpot K. Targeting the tumor microenvironment to enhance antitumor immune responses. Oncotarget 2015; 6:1359-81. [PMID: 25682197 PMCID: PMC4359300 DOI: 10.18632/oncotarget.3204] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 12/24/2014] [Indexed: 12/16/2022] Open
Abstract
The identification of tumor-specific antigens and the immune responses directed against them has instigated the development of therapies to enhance antitumor immune responses. Most of these cancer immunotherapies are administered systemically rather than directly to tumors. Nonetheless, numerous studies have demonstrated that intratumoral therapy is an attractive approach, both for immunization and immunomodulation purposes. Injection, recruitment and/or activation of antigen-presenting cells in the tumor nest have been extensively studied as strategies to cross-prime immune responses. Moreover, delivery of stimulatory cytokines, blockade of inhibitory cytokines and immune checkpoint blockade have been explored to restore immunological fitness at the tumor site. These tumor-targeted therapies have the potential to induce systemic immunity without the toxicity that is often associated with systemic treatments. We review the most promising intratumoral immunotherapies, how these affect systemic antitumor immunity such that disseminated tumor cells are eliminated, and which approaches have been proven successful in animal models and patients.
Collapse
Affiliation(s)
- Kevin Van der Jeught
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Lukasz Bialkowski
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Lidia Daszkiewicz
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Katrijn Broos
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Cleo Goyvaerts
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Dries Renmans
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Sandra Van Lint
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Carlo Heirman
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Kris Thielemans
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| | - Karine Breckpot
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Laarbeeklaan, Jette, Belgium
| |
Collapse
|
142
|
Pellegatta S, Valletta L, Corbetta C, Patanè M, Zucca I, Riccardi Sirtori F, Bruzzone MG, Fogliatto G, Isacchi A, Pollo B, Finocchiaro G. Effective immuno-targeting of the IDH1 mutation R132H in a murine model of intracranial glioma. Acta Neuropathol Commun 2015; 3:4. [PMID: 25849072 PMCID: PMC4359524 DOI: 10.1186/s40478-014-0180-0] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 12/26/2014] [Indexed: 02/07/2023] Open
Abstract
The R132H mutation of cytosolic isocitrate dehydrogenase (IDH1) is present in the majority of low grade gliomas.Immunotherapy in these tumors has an interesting, still unexploited, therapeutic potential, as they are less immunosuppressive than glioblastomas. Using site-directed mutagenesis we introduced the R132H mutation into the murine glioma cell line GL261,creating mIDH1-GL261. Presence of the mutation was confirmed by immunoblotting and production of the oncometabolite 2-hydroxyglutarate (2HG), demonstrated by mass spectrometry (LC-MS/MS) performed on cell supernatant. In vitro mIDH1-GL261 had different morphology but similar growth rate than parental GL261 (p-GL261). After intracranial injection, MRI suggested that the initial growth rate was slower in mIDH1-GL261 than p-GL261 gliomas but overall survival was similar. mIDH1-GL261 gliomas showed evidence of R132H expression and of intratumoral 2HG production (evaluated by MRS and LC-MS/MS). Immunizations were performed nine days after intracranial implantation of mIDH1- or p-GL261 cells by three subcutaneous injections of five different peptides encompassing the IDH1 mutation site, all emulsified with Montanide ISA-51, in association with GM-CSF. Control mice were injected with four ovalbumin peptides or vehicle. Mice with mIDH1-GL261 but not p-GL261 gliomas treated with mIDH1 peptides survived longer than controls; 25% of them were cured. Immunized mice showed higher amounts of peripheral CD8+ T cells, higher production of IFN-γ, and evidence of anti-mIDH1 antibodies.Immunizations led to intratumoral up-regulation of IFN-γ, granzyme-b and perforin-1 and down-regulation of TGF-β2 and IL-10. These results support the translational potential of immunotherapeutic targeting of gliomas carrying IDH1 mutations.
Collapse
|
143
|
Immunobiology and immunotherapeutic targeting of glioma stem cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 853:139-66. [PMID: 25895711 DOI: 10.1007/978-3-319-16537-0_8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
For decades human brain tumors have confounded our efforts to effectively manage and treat patients. In adults, glioblastoma multiforme is the most common malignant brain tumor with a patient survival of just over 14 months. In children, brain tumors are the leading cause of solid tumor cancer death and gliomas account for one-fifth of all childhood cancers. Despite advances in conventional treatments such as surgical resection, radiotherapy, and systemic chemotherapy, the incidence and mortality rates for gliomas have essentially stayed the same. Furthermore, research efforts into novel therapeutics that initially appeared promising have yet to show a marked benefit. A shocking and somewhat disturbing view is that investigators and clinicians may have been targeting the wrong cells, resulting in the appearance of the removal or eradication of patient gliomas only to have brain cancer recurrence. Here we review research progress in immunotherapy as it pertains to glioma treatment and how it can and is being adapted to target glioma stem cells (GSCs) as a means of dealing with this potential paradigm.
Collapse
|
144
|
Calinescu AA, Kamran N, Baker G, Mineharu Y, Lowenstein PR, Castro MG. Overview of current immunotherapeutic strategies for glioma. Immunotherapy 2015; 7:1073-104. [PMID: 26598957 PMCID: PMC4681396 DOI: 10.2217/imt.15.75] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In the last decade, numerous studies of immunotherapy for malignant glioma (glioblastoma multiforme) have brought new knowledge and new hope for improving the prognosis of this incurable disease. Some clinical trials have reached Phase III, following positive outcomes in Phase I and II, with respect to safety and immunological end points. Results are encouraging especially when considering the promise of sustained efficacy by inducing antitumor immunological memory. Progress in understanding the mechanisms of tumor-induced immune suppression led to the development of drugs targeting immunosuppressive checkpoints, which are used in active clinical trials for glioblastoma multiforme. Insights related to the heterogeneity of the disease bring new challenges for the management of glioma and underscore a likely cause of therapeutic failure. An emerging therapeutic strategy is represented by a combinatorial, personalized approach, including the standard of care: surgery, radiation, chemotherapy with added active immunotherapy and multiagent targeting of immunosuppressive checkpoints.
Collapse
Affiliation(s)
| | - Neha Kamran
- Department of Neurosurgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Gregory Baker
- Department of Neurosurgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Yohei Mineharu
- Department of Neurosurgery, Kyoto University, Kyoto, Japan
| | - Pedro Ricardo Lowenstein
- Department of Neurosurgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
- Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Maria Graciela Castro
- Department of Neurosurgery, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
- Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
145
|
Drakes ML, Stiff PJ. Harnessing immunosurveillance: current developments and future directions in cancer immunotherapy. Immunotargets Ther 2014; 3:151-65. [PMID: 27471706 PMCID: PMC4918242 DOI: 10.2147/itt.s37790] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Despite improved methods of cancer detection and disease management over the last few decades, cancer remains a major public health problem in many societies. Conventional therapies, such as chemotherapy, radiation, and surgery, are not usually sufficient to prevent disease recurrence. Therefore, efforts have been focused on developing novel therapies to manage metastatic disease and to prolong disease-free and overall survival, by modulating the immune system to alleviate immunosuppression, and to enhance antitumor immunity. This review discusses protumor mechanisms in patients that circumvent host immunosurveillance, and addresses current immunotherapy modalities designed to target these mechanisms. Given the complexity of cancer immunosuppressive mechanisms, we propose that identification of novel disease biomarkers will drive the development of more targeted immunotherapy. Finally, administration of different classes of immunotherapy in combination regimens, will be the ultimate route to impact low survival rates in advanced cancer patients.
Collapse
Affiliation(s)
- Maureen L Drakes
- Department of Medicine, Division of Hematology and Oncology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Patrick J Stiff
- Department of Medicine, Division of Hematology and Oncology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| |
Collapse
|
146
|
Platten M, Ochs K, Lemke D, Opitz C, Wick W. Microenvironmental clues for glioma immunotherapy. Curr Neurol Neurosci Rep 2014; 14:440. [PMID: 24604058 DOI: 10.1007/s11910-014-0440-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Gliomas have been viewed for decades as inaccessible for a meaningful antitumor immune response as they grow in a sanctuary site protected from infiltrating immune cells. Moreover, the glioma microenvironment constitutes a hostile environment for an efficient antitumor immune response as glioma-derived factors such as transforming growth factor β and catabolites of the essential amino acid tryptophan paralyze T-cell function. There is growing evidence from preclinical and clinical studies that a meaningful antitumor immunity exists in glioma patients and that it can be activated by vaccination strategies. As a consequence, the concept of glioma immunotherapy appears to be experiencing a renaissance with the first phase 3 randomized immunotherapy trials entering the clinical arena. On the basis of encouraging results from other tumor entities using immunostimulatory approaches by blocking endogenous T-cell suppressive pathways mediated by cytotoxic T-lymphocyte antigen 4 or programmed cell death protein 1/programmed cell death protein 1 ligand 1 with humanized antibodies, there is now a realistic and promising option to combine active immunotherapy with agents blocking the immunosuppressive microenvironment in patients with gliomas to allow a peripheral antitumor immune response induced by vaccination to become effective. Here we review the current clinical and preclinical evidence of antimicroenvironment immunotherapeutic strategies in gliomas.
Collapse
Affiliation(s)
- Michael Platten
- Department of Neurooncology, University Hospital Heidelberg and National Center for Tumor Diseases, German Cancer Consortium (DKTK) Clinical Cooperation Units, Im Neuenheimer Feld, Heidelberg, Germany,
| | | | | | | | | |
Collapse
|
147
|
Seemann KM, Luysberg M, Révay Z, Kudejova P, Sanz B, Cassinelli N, Loidl A, Ilicic K, Multhoff G, Schmid TE. Magnetic heating properties and neutron activation of tungsten-oxide coated biocompatible FePt core-shell nanoparticles. J Control Release 2014; 197:131-7. [PMID: 25445697 DOI: 10.1016/j.jconrel.2014.11.007] [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] [Received: 06/13/2014] [Revised: 11/03/2014] [Accepted: 11/04/2014] [Indexed: 10/24/2022]
Abstract
Magnetic nanoparticles are highly desirable for biomedical research and treatment of cancer especially when combined with hyperthermia. The efficacy of nanoparticle-based therapies could be improved by generating radioactive nanoparticles with a convenient decay time and which simultaneously have the capability to be used for locally confined heating. The core-shell morphology of such novel nanoparticles presented in this work involves a polysilico-tungstate molecule of the polyoxometalate family as a precursor coating material, which transforms into an amorphous tungsten oxide coating upon annealing of the FePt core-shell nanoparticles. The content of tungsten atoms in the nanoparticle shell is neutron activated using cold neutrons at the Heinz Maier-Leibnitz (FRMII) neutron facility and thereby transformed into the radioisotope W-187. The sizeable natural abundance of 28% for the W-186 precursor isotope, a radiopharmaceutically advantageous gamma-beta ratio of γβ≈30% and a range of approximately 1mm in biological tissue for the 1.3MeV β-radiation are promising features of the nanoparticles' potential for cancer therapy. Moreover, a high temperature annealing treatment enhances the magnetic moment of nanoparticles in such a way that a magnetic heating effect of several degrees Celsius in liquid suspension - a prerequisite for hyperthermia treatment of cancer - was observed. A rise in temperature of approximately 3°C in aqueous suspension is shown for a moderate nanoparticle concentration of 0.5mg/ml after 15min in an 831kHz high-frequency alternating magnetic field of 250Gauss field strength (25mT). The biocompatibility based on a low cytotoxicity in the non-neutron-activated state in combination with the hydrophilic nature of the tungsten oxide shell makes the coated magnetic FePt nanoparticles ideal candidates for advanced radiopharmaceutical applications.
Collapse
Affiliation(s)
- K M Seemann
- Physik Department E21, Technische Universität München, D-85747 Garching, Germany; Heinz Maier-Leibnitz Zentrum (MLZ), Technische Universität München, D-85747 Garching, Germany.
| | - M Luysberg
- Peter Grünberg Institute (PGI-5) and Ernst-Ruska Centre for Microscopy and Spectroscopy with Electrons, Research Centre Jülich, D-52425 Jülich, Germany
| | - Z Révay
- Heinz Maier-Leibnitz Zentrum (MLZ), Technische Universität München, D-85747 Garching, Germany
| | - P Kudejova
- Heinz Maier-Leibnitz Zentrum (MLZ), Technische Universität München, D-85747 Garching, Germany
| | - B Sanz
- nanoScale Biomagnetics SL, 50012 Zaragoza, Spain
| | - N Cassinelli
- nanoScale Biomagnetics SL, 50012 Zaragoza, Spain
| | - A Loidl
- Experimentalphysik V, Center for Electronic Correlations and Magnetism, Universität Augsburg, D-86159 Augsburg, Germany
| | - K Ilicic
- Klinikum rechts d. Isar, Department of Radiooncology and HMGU München, CCG - Innate Immunity in Tumor Biology", Technische Universität München, D-81675 München, Germany
| | - G Multhoff
- Klinikum rechts d. Isar, Department of Radiooncology and HMGU München, CCG - Innate Immunity in Tumor Biology", Technische Universität München, D-81675 München, Germany
| | - T E Schmid
- Klinikum rechts d. Isar, Department of Radiooncology and HMGU München, CCG - Innate Immunity in Tumor Biology", Technische Universität München, D-81675 München, Germany
| |
Collapse
|
148
|
|
149
|
Krishnan S, Bakker E, Lee C, Kissick HT, Ireland DJ, Beilharz MW. Successful combined intratumoral immunotherapy of established murine mesotheliomas requires B-cell involvement. J Interferon Cytokine Res 2014; 35:100-7. [PMID: 25259549 DOI: 10.1089/jir.2014.0054] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Combination immunotherapy has resulted in a number of impressive outcomes in mouse models and clinical settings. In this study, we report that a timed triple immunotherapy (TTI) protocol using 3 agonist antibodies (anti-CD25mAb, anti-TGF-βmAb, and anti-CTLA-4mAb) produced complete clearance of established AB1 murine mesothelioma tumors. Combining all 3 agonist antibodies into a single cocktail for intratumoral injection was as effective as the TTI in tumor eradication. Cured mice showed elevated levels of tumor-specific IgG antibodies at 95 days posttreatment. Time-course studies of tumor clearance showed (1) that IgG levels were not elevated during tumor clearance and (2) that B-cell numbers were increased in the tumor-draining lymph nodes and spleens during tumor clearance. Finally, employment of B-cell knockout mice indicated a significant role for B cells in the successful eradication of the established tumors by the triple immunotherapy cocktail.
Collapse
Affiliation(s)
- Shruti Krishnan
- 1 School of Pathology and Laboratory Medicine (M502), Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia , Crawley, Australia
| | | | | | | | | | | |
Collapse
|
150
|
New insights into IL-12-mediated tumor suppression. Cell Death Differ 2014; 22:237-46. [PMID: 25190142 DOI: 10.1038/cdd.2014.134] [Citation(s) in RCA: 358] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Revised: 07/17/2014] [Accepted: 07/21/2014] [Indexed: 12/14/2022] Open
Abstract
During the past two decades, interleukin-12 (IL-12) has emerged as one of the most potent cytokines in mediating antitumor activity in a variety of preclinical models. Through pleiotropic effects on different immune cells that form the tumor microenvironment, IL-12 establishes a link between innate and adaptive immunity that involves different immune effector cells and cytokines depending on the type of tumor or the affected tissue. The robust antitumor response exerted by IL-12, however, has not yet been successfully translated into the clinics. The majority of clinical trials involving treatment with IL-12 failed to show sustained antitumor responses and were associated to toxic side effects. Here we discuss the therapeutic effects of IL-12 from preclinical to clinical studies, and will highlight promising strategies to take advantage of the antitumor activity of IL-12 while limiting adverse effects.
Collapse
|