1
|
Rivera D, Bouras A, Mattioli M, Anastasiadou M, Pacentra AC, Pelcher O, Koziel C, Schupper AJ, Chanenchuk T, Carlton H, Ivkov R, Hadjipanayis CG. Magnetic hyperthermia therapy enhances the chemoradiosensitivity of glioblastoma. Sci Rep 2025; 15:10532. [PMID: 40148452 PMCID: PMC11950323 DOI: 10.1038/s41598-025-95544-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/21/2025] [Indexed: 03/29/2025] Open
Abstract
Glioblastoma (GBM) is the most common primary brain cancer and is resistant to standard-of-care chemoradiation therapy (CRT). Magnetic hyperthermia therapy (MHT) exposes magnetic iron oxide nanoparticles (MIONPs) to an alternating magnetic field (AMF) to generate local hyperthermia. This study evaluated MHT-mediated enhancement of CRT in preclinical GBM models. Cell viability and apoptosis were assessed in GBM cell lines after water bath heating with radiation and/or temozolomide. Heating efficiency of MIONPs after intracranial delivery was measured in healthy mice. MHT with CRT was performed in syngeneic and patient-derived xenograft (PDX) GBM tumors. Tissue sections were analyzed for γ-H2AX, HSP90, CD4 + T cells, and microglial cells. Tumor burden and survival were assessed. Hyperthermia with radiation and temozolomide significantly reduced cell viability and increased apoptosis. Hyperthermia predominantly exhibited additive to synergistic interactions with both treatment modalities and reduced doses needed for tumor cell growth inhibition. In vivo, MHT with CRT decreased tumor burden and increased survival in PDX and syngeneic models. Immunohistochemistry showed increased γ-H2AX, HSP90, microglial activation, and CD4 + T cells after MHT in combination with CRT. Overall, adjuvant hyperthermia enhances CRT efficacy in GBM cells, with MHT improving survival outcomes in rodents. Sufficient intracranial heating and MIONP retention for repeated treatments was achieved, supporting further clinical translation.
Collapse
Affiliation(s)
- Daniel Rivera
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Alexandros Bouras
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Milena Mattioli
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
| | - Maria Anastasiadou
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Anna Chiara Pacentra
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
| | - Olivia Pelcher
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
| | - Corrine Koziel
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA
| | - Alexander J Schupper
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Tori Chanenchuk
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Hayden Carlton
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert Ivkov
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Mechanical Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Constantinos G Hadjipanayis
- Brain Tumor Nanotechnology Laboratory, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop Street, Suite F-158, Pittsburgh, PA, 15213, USA.
- Department of Neurological Surgery, Icahn School of Medicine at Mount Sinai, New York City, NY, USA.
| |
Collapse
|
2
|
Erickson NJ, Stavarache M, Tekedereli I, Kaplitt MG, Markert JM. Herpes Simplex Oncolytic Viral Therapy for Malignant Glioma and Mechanisms of Delivery. World Neurosurg 2025; 194:123595. [PMID: 39710201 PMCID: PMC12094189 DOI: 10.1016/j.wneu.2024.123595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/14/2024] [Indexed: 12/24/2024]
Abstract
The authors present a comprehensive review on the history and development of oncolytic herpes simplex viral therapies for malignant glioma with a focus on mechanisms of delivery in prior and ongoing clinical trials. This review highlights the advancements made with regard to delivering these therapies to a highly complex immunologic environment in the setting of the blood-brain and blood-tumor barrier in a safe and effective manner.
Collapse
Affiliation(s)
- Nicholas J Erickson
- Department of Neurosurgery, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mihaela Stavarache
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medicine, New York, New York, USA
| | - Ibrahim Tekedereli
- Department of Neurosurgery, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Michael G Kaplitt
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medicine, New York, New York, USA
| | - James M Markert
- Department of Neurosurgery, The University of Alabama at Birmingham, Birmingham, Alabama, USA.
| |
Collapse
|
3
|
Basina G, Diamantopoulos G, Devlin E, Psycharis V, Alhassan SM, Pissas M, Hadjipanayis G, Tomou A, Bouras A, Hadjipanayis C, Tzitzios V. LAPONITE® nanodisk-"decorated" Fe 3O 4 nanoparticles: a biocompatible nano-hybrid with ultrafast magnetic hyperthermia and MRI contrast agent ability. J Mater Chem B 2022; 10:4935-4943. [PMID: 35535802 DOI: 10.1039/d2tb00139j] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Magnetic Fe3O4 nanoparticles "decorated" by LAPONITE® nanodisks have been materialized utilizing the Schikorr reaction following a facile approach and tested as mediators of heat for localized magnetic hyperthermia (MH) and as magnetic resonance imaging (MRI) agents. The synthetic protocol involves the interaction between two layered inorganic compounds, ferrous hydroxide, Fe(OH)2, and the synthetic smectite LAPONITE® clay Na0.7+[(Si8Mg5.5Li0.3)O20(OH)4]0.7-, towards the formation of superparamagnetic Fe3O4 nanoparticles, which are well decorated by the diamagnetic clay nanodisks. The latter imparts high negative ζ-potential values (up to -34.1 mV) to the particles, which provide stability against flocculation and precipitation, resulting in stable water dispersions. The obtained LAPONITE®-"decorated" Fe3O4 nanohybrids were characterized by powder X-ray diffraction (XRD), transmission electron microscopy (TEM), Mössbauer spectroscopy, dynamic light scattering (DLS) and vibrating sample magnetometry (VSM) at room temperature, revealing superior magnetic hyperthermia performance with specific absorption rate (SAR) values reaching 540 W gFe-1 (28 kA m-1, 150 kHz) for the hybrid material with a magnetic loading of 50 wt% Fe3O4/LAPONITE®. Toxicity studies were also performed with human glioblastoma (GBM) cells and human foreskin fibroblasts (HFF), which show negligible to no toxicity. Furthermore, T2-weighted MR imaging of rodent brain shows that the LAPONITE®-"decorated" Fe3O4 nanohybrids predominantly affected the transverse T2 relaxation time of tissue water, which resulted in a signal drop on the MRI T2-weighted imaging, allowing for imaging of the magnetic nanoparticles.
Collapse
Affiliation(s)
- Georgia Basina
- Department of Physics and Astronomy, University of Delaware, Newark, DE 19711, USA. .,Institute of Nanoscience and Nanotechnology, NCSR Demokritos, 15310, Athens, Greece.
| | - George Diamantopoulos
- Institute of Nanoscience and Nanotechnology, NCSR Demokritos, 15310, Athens, Greece.
| | - Eamonn Devlin
- Institute of Nanoscience and Nanotechnology, NCSR Demokritos, 15310, Athens, Greece.
| | - Vassilis Psycharis
- Institute of Nanoscience and Nanotechnology, NCSR Demokritos, 15310, Athens, Greece.
| | - Saeed M Alhassan
- Department of Chemical Engineering, Khalifa University, P.O. Box 127788, Abu Dhabi, United Arab Emirates
| | - Michael Pissas
- Institute of Nanoscience and Nanotechnology, NCSR Demokritos, 15310, Athens, Greece.
| | - George Hadjipanayis
- Department of Physics and Astronomy, University of Delaware, Newark, DE 19711, USA.
| | - Aphrodite Tomou
- Institute of Nanoscience and Nanotechnology, NCSR Demokritos, 15310, Athens, Greece. .,Goodfellow Cambridge Ltd., Ermine Business Park, Huntingdon PE29 6WR, Cambridge, UK
| | - Alexandros Bouras
- Brain Tumor Nanotechnology Laboratory, Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Constantinos Hadjipanayis
- Brain Tumor Nanotechnology Laboratory, Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Vasileios Tzitzios
- Institute of Nanoscience and Nanotechnology, NCSR Demokritos, 15310, Athens, Greece. .,Department of Chemical Engineering, Khalifa University, P.O. Box 127788, Abu Dhabi, United Arab Emirates
| |
Collapse
|
4
|
Janjua TI, Rewatkar P, Ahmed-Cox A, Saeed I, Mansfeld FM, Kulshreshtha R, Kumeria T, Ziegler DS, Kavallaris M, Mazzieri R, Popat A. Frontiers in the treatment of glioblastoma: Past, present and emerging. Adv Drug Deliv Rev 2021; 171:108-138. [PMID: 33486006 DOI: 10.1016/j.addr.2021.01.012] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/13/2020] [Accepted: 01/09/2021] [Indexed: 12/13/2022]
Abstract
Glioblastoma (GBM) is one of the most aggressive cancers of the brain. Despite extensive research over the last several decades, the survival rates for GBM have not improved and prognosis remains poor. To date, only a few therapies are approved for the treatment of GBM with the main reasons being: 1) significant tumour heterogeneity which promotes the selection of resistant subpopulations 2) GBM induced immunosuppression and 3) fortified location of the tumour in the brain which hinders the delivery of therapeutics. Existing therapies for GBM such as radiotherapy, surgery and chemotherapy have been unable to reach the clinical efficacy necessary to prolong patient survival more than a few months. This comprehensive review evaluates the current and emerging therapies including those in clinical trials that may potentially improve both targeted delivery of therapeutics directly to the tumour site and the development of agents that may specifically target GBM. Particular focus has also been given to emerging delivery technologies such as focused ultrasound, cellular delivery systems nanomedicines and immunotherapy. Finally, we discuss the importance of developing novel materials for improved delivery efficacy of nanoparticles and therapeutics to reduce the suffering of GBM patients.
Collapse
|
5
|
Loya J, Zhang C, Cox E, Achrol AS, Kesari S. Biological intratumoral therapy for the high-grade glioma part II: vector- and cell-based therapies and radioimmunotherapy. CNS Oncol 2019; 8:CNS40. [PMID: 31747784 PMCID: PMC6880300 DOI: 10.2217/cns-2019-0002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Management of high-grade gliomas (HGGs) remains a complex challenge with an overall poor prognosis despite aggressive multimodal treatment. New translational research has focused on maximizing tumor cell eradication through improved tumor cell targeting while minimizing collateral systemic side effects. In particular, biological intratumoral therapies have been the focus of novel translational research efforts due to their inherent potential to be both dynamically adaptive and target specific. This two part review will provide an overview of biological intratumoral therapies that have been evaluated in human clinical trials in HGGs, and summarize key advances and remaining challenges in the development of these therapies as a potential new paradigm in the management of HGGs. Part II discusses vector-based therapies, cell-based therapies and radioimmunotherapy.
Collapse
Affiliation(s)
- Joshua Loya
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Charlie Zhang
- Buffalo School of Medicine, State University of New York, Buffalo, NY 14202, USA
| | - Emily Cox
- Providence Medical Research Center, Spokane, WA 99204, USA
| | - Achal S Achrol
- John Wayne Cancer Institute, Pacific Neuroscience Institute, Santa Monica, CA 90404, USA
| | - Santosh Kesari
- John Wayne Cancer Institute, Pacific Neuroscience Institute, Santa Monica, CA 90404, USA
| |
Collapse
|
6
|
Lynes J, Sanchez V, Dominah G, Nwankwo A, Nduom E. Current Options and Future Directions in Immune Therapy for Glioblastoma. Front Oncol 2018; 8:578. [PMID: 30568917 PMCID: PMC6290347 DOI: 10.3389/fonc.2018.00578] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 11/19/2018] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma is in need of innovative treatment approaches. Immune therapy for cancer refers to the use of the body's immune system to target malignant cells in the body. Such immune therapeutics have recently been very successful in treating a diverse group of cancerous lesions. As a result, many new immune therapies have gained Food and Drug Administration approval for the treatment of cancer, and there has been an explosion in the study of immune therapeutics for cancer treatment over the past few years. However, the immune suppression of glioblastoma and the unique immune microenvironment of the brain make immune therapeutics more challenging to apply to the brain than to other systemic cancers. Here, we discuss the existing barriers to successful immune therapy for glioblastoma and the ongoing development of immune therapeutics. We will discuss the discovery and classification of immune suppressive factors in the glioblastoma microenvironment; the development of vaccine-based therapies; the use of convection-enhanced delivery to introduce tumoricidal viruses into the tumor microenvironment, leading to secondary immune responses; the emerging use of adoptive cell therapy in the treatment of glioblastoma; and future frontiers, such as the use of cerebral microdialysis for immune monitoring and the use of sequencing to develop patient-specific therapeutics. Armed with a better understanding of the challenges inherent in immune therapy for glioblastoma, we may soon see more successes in immune-based clinical trials for this deadly disease.
Collapse
Affiliation(s)
- John Lynes
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, United States.,MedStar Georgetown University Hospital, Washington, DC, United States
| | - Victoria Sanchez
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, United States
| | - Gifty Dominah
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, United States
| | - Anthony Nwankwo
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, United States
| | - Edjah Nduom
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, United States
| |
Collapse
|
7
|
Unlocking the promise of oncolytic virotherapy in glioma: combination with chemotherapy to enhance efficacy. Ther Deliv 2016; 6:453-68. [PMID: 25996044 DOI: 10.4155/tde.14.123] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Malignant glioma is a relentless burden to both patients and clinicians, and calls for innovation to overcome the limitations in current management. Glioma therapy using viruses has been investigated to accentuate the nature of a virus, killing a host tumor cell during its replication. As virus mediated approaches progress with promising therapeutic advantages, combination therapy with chemotherapy and oncolytic viruses has emerged as a more synergistic and possibly efficacious therapy. Here, we will review malignant glioma as well as prior experience with oncolytic viruses, chemotherapy and combination of the two, examining how the combination can be optimized in the future.
Collapse
|
8
|
Liu H, Zhang J, Chen X, Du XS, Zhang JL, Liu G, Zhang WG. Application of iron oxide nanoparticles in glioma imaging and therapy: from bench to bedside. NANOSCALE 2016; 8:7808-7826. [PMID: 27029509 DOI: 10.1039/c6nr00147e] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Gliomas are the most common primary brain tumors and have a very dismal prognosis. However, recent advancements in nanomedicine and nanotechnology provide opportunities for personalized treatment regimens to improve the poor prognosis of patients suffering from glioma. This comprehensive review starts with an outline of the current status facing glioma. It then provides an overview of the state-of-the-art applications of iron oxide nanoparticles (IONPs) to glioma diagnostics and therapeutics, including MR contrast enhancement, drug delivery, cell labeling and tracking, magnetic hyperthermia treatment and magnetic particle imaging. It also addresses current challenges associated with the biological barriers and IONP design with an emphasis on recent advances and innovative approaches for glioma targeting strategies. Opportunities for future development are highlighted.
Collapse
Affiliation(s)
- Heng Liu
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China and State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China.
| | - Jun Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China. and Sichuan Key Laboratory of Medical Imaging, Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong 637007, China
| | - Xiao Chen
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Xue-Song Du
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Jin-Long Zhang
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China.
| | - Wei-Guo Zhang
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China and The State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
| |
Collapse
|
9
|
Glasgow MDK, Chougule MB. Recent Developments in Active Tumor Targeted Multifunctional Nanoparticles for Combination Chemotherapy in Cancer Treatment and Imaging. J Biomed Nanotechnol 2016; 11:1859-98. [PMID: 26554150 DOI: 10.1166/jbn.2015.2145] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nanotechnology and combination therapy are two major fields that show great promise in the treatment of cancer. The delivery of drugs via nanoparticles helps to improve drug's therapeutic effectiveness while reducing adverse side effects associated wifh high dosage by improving their pharmacokinetics. Taking advantage of molecular markers over-expressing on tumor tissues compared to normal cells, an "active" molecular marker targeted approach would be-beneficial for cancer therapy. These actively targeted nanoparticles would increase drug concentration at the tumor site, improving efficacy while further reducing chemo-resistance. The multidisciplinary approach may help to improve the overall efficacy in cancer therapy. This review article summarizes recent developments of targeted multifunctional nanoparticles in the delivery, of various drugs for a combinational chemotherapy approach to cancer treatment and imaging.
Collapse
|
10
|
Pourgholi F, Hajivalili M, Farhad JN, Kafil HS, Yousefi M. Nanoparticles: Novel vehicles in treatment of Glioblastoma. Biomed Pharmacother 2015; 77:98-107. [PMID: 26796272 DOI: 10.1016/j.biopha.2015.12.014] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 12/06/2015] [Accepted: 12/15/2015] [Indexed: 01/05/2023] Open
Abstract
Glioblastoma multiform (GBM) is the most common brain tumor. The current GBM treatments comprise of radiation therapy, chemotherapy and surgery. One of the most important problems regarding the treatment of GBM is the presence of blood brain barrier (BBB) which inhibits the efficient drug delivery into central nervous system (CNS). Nanothechnology can help to deliver therapeutic drugs into CNS through crossing the BBB. There are different types of nanoparticles (Nps) which can be manipulated for clinical applications as a treatment for CNS-related disorders. In this review, we will discuss the role of Nps in the treatment of GBM.
Collapse
Affiliation(s)
- Fatemeh Pourgholi
- Tuberculosis and Lung Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahsa Hajivalili
- Tuberculosis and Lung Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jadidi-Niaragh Farhad
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Samadi Kafil
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
11
|
Bouras A, Kaluzova M, Hadjipanayis CG. Radiosensitivity enhancement of radioresistant glioblastoma by epidermal growth factor receptor antibody-conjugated iron-oxide nanoparticles. J Neurooncol 2015; 124:13-22. [PMID: 25981803 DOI: 10.1007/s11060-015-1807-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 05/08/2015] [Indexed: 01/04/2023]
Abstract
The epidermal growth factor receptor deletion variant EGFRvIII is known to be expressed in a subset of patients with glioblastoma (GBM) tumors that enhances tumorigenicity and also accounts for radiation and chemotherapy resistance. Targeting the EGFRvIII deletion mutant may lead to improved GBM therapy and better patient prognosis. Multifunctional magnetic nanoparticles serve as a potential clinical tool that can provide cancer cell targeted drug delivery, imaging, and therapy. Our previous studies have shown that an EGFRvIII-specific antibody and cetuximab (an EGFR- and EGFRvIII-specific antibody), when bioconjugated to IONPs (EGFRvIII-IONPs or cetuximab-IONPs respectively), can simultaneously provide sensitive cancer cell detection by magnetic resonance imaging (MRI) and targeted therapy of experimental GBM. In this study, we investigated whether cetuximab-IONPs can additionally allow for the radiosensitivity enhancement of GBM. Cetuximab-IONPs were used in combination with single (10 Gy × 1) or multiple fractions (10 Gy × 2) of ionizing radiation (IR) for radiosensitization of EGFRvIII-overexpressing human GBM cells in vitro and in vivo after convection-enhanced delivery (CED). A significant GBM antitumor effect was observed in vitro after treatment with cetuximab-IONPs and subsequent single or fractionated IR. A significant increase in overall survival of nude mice implanted with human GBM xenografts was found after treatment by cetuximab-IONP CED and subsequent fractionated IR. Increased DNA double strands breaks (DSBs), as well as increased reactive oxygen species (ROS) formation, were felt to represent the mediators of the observed radiosensitization effect with the combination therapy of IR and cetuximab-IONPs treatment.
Collapse
Affiliation(s)
- Alexandros Bouras
- Brain Tumor Nanotechnology Laboratory, Department of Neurosurgery, Emory University School of Medicine, Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA
| | | | | |
Collapse
|
12
|
Clarke RH, Moosa S, Anzivino M, Wang Y, Floyd DH, Purow BW, Lee KS. Sustained radiosensitization of hypoxic glioma cells after oxygen pretreatment in an animal model of glioblastoma and in vitro models of tumor hypoxia. PLoS One 2014; 9:e111199. [PMID: 25350400 PMCID: PMC4211739 DOI: 10.1371/journal.pone.0111199] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 09/29/2014] [Indexed: 12/31/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and lethal form of brain cancer and these tumors are highly resistant to chemo- and radiotherapy. Radioresistance is thought to result from a paucity of molecular oxygen in hypoxic tumor regions, resulting in reduced DNA damage and enhanced cellular defense mechanisms. Efforts to counteract tumor hypoxia during radiotherapy are limited by an attendant increase in the sensitivity of healthy brain tissue to radiation. However, the presence of heightened levels of molecular oxygen during radiotherapy, while conventionally deemed critical for adjuvant oxygen therapy to sensitize hypoxic tumor tissue, might not actually be necessary. We evaluated the concept that pre-treating tumor tissue by transiently elevating tissue oxygenation prior to radiation exposure could increase the efficacy of radiotherapy, even when radiotherapy is administered after the return of tumor tissue oxygen to hypoxic baseline levels. Using nude mice bearing intracranial U87-luciferase xenografts, and in vitro models of tumor hypoxia, the efficacy of oxygen pretreatment for producing radiosensitization was tested. Oxygen-induced radiosensitization of tumor tissue was observed in GBM xenografts, as seen by suppression of tumor growth and increased survival. Additionally, rodent and human glioma cells, and human glioma stem cells, exhibited prolonged enhanced vulnerability to radiation after oxygen pretreatment in vitro, even when radiation was delivered under hypoxic conditions. Over-expression of HIF-1α reduced this radiosensitization, indicating that this effect is mediated, in part, via a change in HIF-1-dependent mechanisms. Importantly, an identical duration of transient hyperoxic exposure does not sensitize normal human astrocytes to radiation in vitro. Taken together, these results indicate that briefly pre-treating tumors with elevated levels of oxygen prior to radiotherapy may represent a means for selectively targeting radiation-resistant hypoxic cancer cells, and could serve as a safe and effective adjuvant to radiation therapy for patients with GBM.
Collapse
Affiliation(s)
- Ryon H. Clarke
- Department of Neuroscience, University of Virginia, Charlottesville, VA, United States of America
- School of Medicine, University of Virginia Health System, Charlottesville, VA, United States of America
| | - Shayan Moosa
- School of Medicine, University of Virginia Health System, Charlottesville, VA, United States of America
| | - Matthew Anzivino
- Department of Neuroscience, University of Virginia, Charlottesville, VA, United States of America
- School of Medicine, University of Virginia Health System, Charlottesville, VA, United States of America
| | - Yi Wang
- Department of Neuroscience, University of Virginia, Charlottesville, VA, United States of America
- School of Medicine, University of Virginia Health System, Charlottesville, VA, United States of America
| | - Desiree Hunt Floyd
- School of Medicine, University of Virginia Health System, Charlottesville, VA, United States of America
- Division of Neuro-Oncology, Departments of Neurology, Microbiology, and Biochemistry and Molecular Genetics, University of Virginia Health System, Charlottesville, VA, United States of America
| | - Benjamin W. Purow
- School of Medicine, University of Virginia Health System, Charlottesville, VA, United States of America
- Division of Neuro-Oncology, Departments of Neurology, Microbiology, and Biochemistry and Molecular Genetics, University of Virginia Health System, Charlottesville, VA, United States of America
| | - Kevin S. Lee
- Department of Neuroscience, University of Virginia, Charlottesville, VA, United States of America
- School of Medicine, University of Virginia Health System, Charlottesville, VA, United States of America
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, United States of America
| |
Collapse
|
13
|
Ning J, Wakimoto H. Oncolytic herpes simplex virus-based strategies: toward a breakthrough in glioblastoma therapy. Front Microbiol 2014; 5:303. [PMID: 24999342 PMCID: PMC4064532 DOI: 10.3389/fmicb.2014.00303] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/03/2014] [Indexed: 12/12/2022] Open
Abstract
Oncolytic viruses (OV) are a class of antitumor agents that selectively kill tumor cells while sparing normal cells. Oncolytic herpes simplex virus (oHSV) has been investigated in clinical trials for patients with the malignant brain tumor glioblastoma for more than a decade. These clinical studies have shown the safety of oHSV administration to the human brain, however, therapeutic efficacy of oHSV as a single treatment remains unsatisfactory. Factors that could hamper the anti-glioblastoma efficacy of oHSV include: attenuated potency of oHSV due to deletion or mutation of viral genes involved in virulence, restricting viral replication and spread within the tumor; suboptimal oHSV delivery associated with intratumoral injection; virus infection-induced inflammatory and cellular immune responses which could inhibit oHSV replication and promote its clearance; lack of effective incorporation of oHSV into standard-of-care, and poor knowledge about the ability of oHSV to target glioblastoma stem cells (GSCs). In an attempt to address these issues, recent research efforts have been directed at: (1) design of new engineered viruses to enhance potency, (2) better understanding of the role of the cellular immunity elicited by oHSV infection of tumors, (3) combinatorial strategies with different antitumor agents with a mechanistic rationale, (4) “armed” viruses expressing therapeutic transgenes, (5) use of GSC-derived models in oHSV evaluation, and (6) combinations of these. In this review, we will describe the current status of oHSV clinical trials for glioblastoma, and discuss recent research advances and future directions toward successful oHSV-based therapy of glioblastoma.
Collapse
Affiliation(s)
- Jianfang Ning
- Department of Neurosurgery, Brain Tumor Research Center, Massachusetts General Hospital, Harvard Medical School Boston, MA, USA
| | - Hiroaki Wakimoto
- Department of Neurosurgery, Brain Tumor Research Center, Massachusetts General Hospital, Harvard Medical School Boston, MA, USA
| |
Collapse
|
14
|
Braidwood L, Graham SV, Graham A, Conner J. Oncolytic herpes viruses, chemotherapeutics, and other cancer drugs. Oncolytic Virother 2013; 2:57-74. [PMID: 27512658 PMCID: PMC4918355 DOI: 10.2147/ov.s52601] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Oncolytic viruses are emerging as a potential new way of treating cancers. They are selectively replication-competent viruses that propagate only in actively dividing tumor cells but not in normal cells and, as a result, destroy the tumor cells by consequence of lytic infection. At least six different oncolytic herpes simplex viruses (oHSVs) have undergone clinical trials worldwide to date, and they have demonstrated an excellent safety profile and intimations of efficacy. The first pivotal Phase III trial with an oHSV, talimogene laherparepvec (T-Vec [OncoVex(GM-CSF)]), is almost complete, with extremely positive early results reported. Intuitively, therapeutically beneficial interactions between oHSV and chemotherapeutic and targeted therapeutic drugs would be limited as the virus requires actively dividing cells for maximum replication efficiency and most anticancer agents are cytotoxic or cytostatic. However, combinations of such agents display a range of responses, with antagonistic, additive, or, perhaps most surprisingly, synergistic enhancement of antitumor activity. When synergistic interactions in cancer cell killing are observed, chemotherapy dose reductions that achieve the same overall efficacy may be possible, resulting in a valuable reduction of adverse side effects. Therefore, the combination of an oHSV with "standard-of-care" drugs makes a logical and reasonable approach to improved therapy, and the addition of a targeted oncolytic therapy with "standard-of-care" drugs merits further investigation, both preclinically and in the clinic. Numerous publications report such studies of oncolytic HSV in combination with other drugs, and we review their findings here. Viral interactions with cellular hosts are complex and frequently involve intracellular signaling networks, thus creating diverse opportunities for synergistic or additive combinations with many anticancer drugs. We discuss potential mechanisms that may lead to synergistic interactions.
Collapse
Affiliation(s)
- Lynne Braidwood
- Virttu Biologics Ltd, Department of Neurology, Southern General Hospital, Glasgow, UK
| | - Sheila V Graham
- MRC-University of Glasgow Centre for Virus Research, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, Jarrett Building, University of Glasgow, Glasgow, UK
| | - Alex Graham
- Virttu Biologics Ltd, Department of Neurology, Southern General Hospital, Glasgow, UK
| | - Joe Conner
- Virttu Biologics Ltd, Department of Neurology, Southern General Hospital, Glasgow, UK
| |
Collapse
|
15
|
The clinical potential of targeted nanomedicine: delivering to cancer stem-like cells. Mol Ther 2013; 22:278-291. [PMID: 24113515 DOI: 10.1038/mt.2013.231] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 09/19/2013] [Indexed: 12/18/2022] Open
Abstract
Cancer stem-like cells (CSCs) have been implicated in recurrence and treatment resistance in many human cancers. Thus, a CSC-targeted drug delivery strategy to eliminate CSCs is a desirable approach for developing a more effective anticancer therapy. We have developed a tumor-targeting nanodelivery platform (scL) for systemic administration of molecular medicines. Following treatment with the scL nanocomplex carrying various payloads, we have observed exquisite tumor-targeting specificity and significant antitumor response with long-term survival benefit in numerous animal models. We hypothesized that this observed efficacy might be attributed, at least in part, to elimination of CSCs. Here, we demonstrate the ability of scL to target both CSCs and differentiated nonstem cancer cells (non-CSCs) in various mouse models including subcutaneous and intracranial xenografts, syngeneic, and chemically induced tumors. We also show that systemic administration of scL carrying the wtp53 gene was able to induce tumor growth inhibition and the death of both CSCs and non-CSCs in subcutaneous colorectal cancer xenografts suggesting that this could be an effective method to reduce cancer recurrence and treatment resistance. This scL nanocomplex is being evaluated in a number of clinical trials where it has been shown to be well tolerated with indications of anticancer activity.
Collapse
|
16
|
Salehi A, Chen CC, Carter BS. Viral vectors: promising new therapeutics in the battle against glioblastoma. Expert Rev Clin Pharmacol 2012; 5:489-91. [PMID: 23121267 DOI: 10.1586/ecp.12.43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
17
|
Miladi I, Duc GL, Kryza D, Berniard A, Mowat P, Roux S, Taleb J, Bonazza P, Perriat P, Lux F, Tillement O, Billotey C, Janier M. Biodistribution of ultra small gadolinium-based nanoparticles as theranostic agent: Application to brain tumors. J Biomater Appl 2012; 28:385-94. [DOI: 10.1177/0885328212454315] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Gadolinium-based nanoparticles are novel objects with interesting physical properties, allowing their use for diagnostic and therapeutic applications. Gadolinium-based nanoparticles were imaged following intravenous injection in healthy rats and rats grafted with 9L gliosarcoma tumors using magnetic resonance imaging and scintigraphic imaging. Quantitative biodistribution using gamma-counting of each sampled organ confirmed that these nanoparticles were rapidly cleared essentially by renal excretion. Accumulation of these nanoparticles in 9L gliosarcoma tumors implanted in the rat brain was quantitated. This passive and long-duration accumulation of gadolinium-based nanoparticles in tumor, which is related to disruption of the blood–brain barrier, is in good agreement with the use of these nanoparticles as radiosensitizers for brain tumors.
Collapse
Affiliation(s)
- Imen Miladi
- Laboratoire de Physico-Chimie des Matériaux Luminescents, UMR 5620 CNRS, Université Claude Bernard Lyon 1, Villeurbanne Cedex, France
| | - Géraldine Le Duc
- European Synchrotron Radiation Facility, ID 17 Biomedical Beamline, Polygone Scientifique Louis Néel, Grenoble, France
| | - David Kryza
- Laboratoire de Physico-Chimie des Matériaux Luminescents, UMR 5620 CNRS, Université Claude Bernard Lyon 1, Villeurbanne Cedex, France
- Hospices Civils de Lyon, Service de Médecine Nucléaire, Hôpital Edouard Herriot, Lyon, France
| | - Aurélie Berniard
- Laboratoire de Physico-Chimie des Matériaux Luminescents, UMR 5620 CNRS, Université Claude Bernard Lyon 1, Villeurbanne Cedex, France
| | - Pierre Mowat
- Laboratoire de Physico-Chimie des Matériaux Luminescents, UMR 5620 CNRS, Université Claude Bernard Lyon 1, Villeurbanne Cedex, France
| | - Stéphane Roux
- Laboratoire de Physico-Chimie des Matériaux Luminescents, UMR 5620 CNRS, Université Claude Bernard Lyon 1, Villeurbanne Cedex, France
- Institut UTINAM, UMR 6213 CNRS, Université de Franche-Comté, Besançon Cedex, France
| | - Jacqueline Taleb
- Laboratoire de Physico-Chimie des Matériaux Luminescents, UMR 5620 CNRS, Université Claude Bernard Lyon 1, Villeurbanne Cedex, France
| | - Pauline Bonazza
- Laboratoire de Physico-Chimie des Matériaux Luminescents, UMR 5620 CNRS, Université Claude Bernard Lyon 1, Villeurbanne Cedex, France
| | - Pascal Perriat
- Matériaux Ingénierie et Science, UMR 5510 CNRS, Villeurbanne Cedex, France
| | - François Lux
- Laboratoire de Physico-Chimie des Matériaux Luminescents, UMR 5620 CNRS, Université Claude Bernard Lyon 1, Villeurbanne Cedex, France
| | - Olivier Tillement
- Laboratoire de Physico-Chimie des Matériaux Luminescents, UMR 5620 CNRS, Université Claude Bernard Lyon 1, Villeurbanne Cedex, France
| | - Claire Billotey
- Laboratoire de Physico-Chimie des Matériaux Luminescents, UMR 5620 CNRS, Université Claude Bernard Lyon 1, Villeurbanne Cedex, France
- Hospices Civils de Lyon, Service de Médecine Nucléaire, Hôpital Edouard Herriot, Lyon, France
| | - Marc Janier
- Laboratoire de Physico-Chimie des Matériaux Luminescents, UMR 5620 CNRS, Université Claude Bernard Lyon 1, Villeurbanne Cedex, France
- Hospices Civils de Lyon, Service de Médecine Nucléaire, Hôpital Edouard Herriot, Lyon, France
| |
Collapse
|
18
|
Yu MK, Park J, Jon S. Targeting strategies for multifunctional nanoparticles in cancer imaging and therapy. Theranostics 2012; 2:3-44. [PMID: 22272217 PMCID: PMC3263514 DOI: 10.7150/thno.3463] [Citation(s) in RCA: 547] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 09/28/2011] [Indexed: 12/11/2022] Open
Abstract
Nanomaterials offer new opportunities for cancer diagnosis and treatment. Multifunctional nanoparticles harboring various functions including targeting, imaging, therapy, and etc have been intensively studied aiming to overcome limitations associated with conventional cancer diagnosis and therapy. Of various nanoparticles, magnetic iron oxide nanoparticles with superparamagnetic property have shown potential as multifunctional nanoparticles for clinical translation because they have been used asmagnetic resonance imaging (MRI) constrast agents in clinic and their features could be easily tailored by including targeting moieties, fluorescence dyes, or therapeutic agents. This review summarizes targeting strategies for construction of multifunctional nanoparticles including magnetic nanoparticles-based theranostic systems, and the various surface engineering strategies of nanoparticles for in vivo applications.
Collapse
Affiliation(s)
| | | | - Sangyong Jon
- Cell Dynamics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, 261 Chemdangwagi-ro, Gwangju 500-712, Republic of Korea
| |
Collapse
|
19
|
Yu MK, Park J, Jon S. Magnetic nanoparticles and their applications in image-guided drug delivery. Drug Deliv Transl Res 2011; 2:3-21. [DOI: 10.1007/s13346-011-0049-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
20
|
Muik A, von Laer D. Oncolytic virotherapy of glioma: what does it need to make it work? Future Virol 2011. [DOI: 10.2217/fvl.11.111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Oncolytic virotherapy is a highly promising approach, with diverse viruses currently under development as therapeutic agents to treat malignant glioma. Although the first clinical trials did not show toxicity or serious adverse events related to intracerebral administration, overall the antitumor efficacy has fallen short of expectations. This article discusses multiple options on how to improve and maximize the effectiveness of oncolytic virus therapy in brain cancer, including strategies to enhance safety by attenuating neurovirulence via cancer-specific cell-targeting, increasing antitumor potency by transgene-arming and integrating the ability to trigger an effective antitumoral immune response, as well as developing optimized delivery routes in order to boost intratumoral viral distribution. Eventually, it will highlight the use of multipronged approaches, combining multiple modes of action of different agents.
Collapse
Affiliation(s)
| | - Dorothee von Laer
- Division of Virology, Innsbruck Medical University, Fritz-Pregl-Str. 3, A-6020 Innsbruck, Austria
| |
Collapse
|
21
|
Touchefeu Y, Vassaux G, Harrington KJ. Oncolytic viruses in radiation oncology. Radiother Oncol 2011; 99:262-70. [PMID: 21704402 DOI: 10.1016/j.radonc.2011.05.078] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Revised: 05/27/2011] [Accepted: 05/28/2011] [Indexed: 10/18/2022]
Abstract
Oncolytic viruses are investigational cancer treatments. They are currently being assessed as single agents or in combination with standard therapies such as external beam radiotherapy - a DNA damaging agent that is a standard of care for many tumour types. Preclinical data indicate that combinations of oncolytic viruses and radiation therapy are promising, showing additional or synergistic antitumour effects in in vitro and in vivo studies. This interaction has the potential to be multifaceted: viruses may act as radiosensitizing agents, but radiation may also enhance viral oncolysis by increasing viral uptake, replication, gene expression and cell death (apoptosis, autophagy or necrosis) in irradiated cells. Phase I and II clinical trials investigating combinations of viruses and radiation therapy have been completed, paving the way for ongoing phase III studies. The aim of this review is to focus on the therapeutic potential of these combinations and to highlight their mechanistic bases, with particular emphasis on the role of the DNA damage response.
Collapse
Affiliation(s)
- Yann Touchefeu
- The Institute of Cancer Research, Section of Cell and Molecular Biology, London, UK
| | | | | |
Collapse
|
22
|
Smith E, Breznik J, Lichty BD. Strategies to enhance viral penetration of solid tumors. Hum Gene Ther 2011; 22:1053-60. [PMID: 21443415 DOI: 10.1089/hum.2010.227] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The efficient delivery of viral vectors to tumors is an active area of investigation. A number of barriers exist that must be overcome to achieve good penetration of vectors into tumors and distribution of their effects throughout the tumor mass. Replicating oncolytic viruses have the advantage of being able to amplify the initial dose, but progeny virus are prevented from spreading because of a dense mass of tightly packed cells with a dense extracellular matrix, admixed normal stromal cells, and high interstitial pressure. Although intratumoral injection may ensure initial delivery the distribution achieved by intravenous administration may be superior and come with beneficial bystander damage to the tumor vasculature. Strategies to enhance intravenous delivery and subsequent spread of these vectors within tumors are being developed by a number of groups. Achieving the goal of efficient penetration and spread of viruses within solid tumors is a necessary prerequisite to significant improvements in virus-vectored therapy of solid tumors.
Collapse
Affiliation(s)
- Elspeth Smith
- Centre for Gene Therapeutics, McMaster University, Hamilton, ON, Canada L8N 3Z5
| | | | | |
Collapse
|
23
|
Kaestle C, Winkeler A, Richter R, Sauer H, Hescheler J, Fraefel C, Wartenberg M, Jacobs AH. Imaging Herpes Simplex Virus Type 1 Amplicon Vector–Mediated Gene Expression in Human Glioma Spheroids. Mol Imaging 2011; 10:197-205. [DOI: 10.2310/7290.2010.00036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2010] [Accepted: 03/24/2010] [Indexed: 11/18/2022] Open
Affiliation(s)
- Christine Kaestle
- From the Laboratory for Gene Therapy and Molecular Imaging at the Max Planck Institute for Neurological Research, Cologne, Germany; Department of Physiology, Faculty of Medicine, Justus Liebig University, Giessen, Germany; Institute for Neurophysiology, University of Cologne, Cologne, Germany; Institute of Virology, University of Zurich, Zurich, Switzerland; Cardiology Division, Clinic of Internal Medicine I, Friedrich Schiller University, Jena, Germany; and European Institute for Molecular Imaging,
| | - Alexandra Winkeler
- From the Laboratory for Gene Therapy and Molecular Imaging at the Max Planck Institute for Neurological Research, Cologne, Germany; Department of Physiology, Faculty of Medicine, Justus Liebig University, Giessen, Germany; Institute for Neurophysiology, University of Cologne, Cologne, Germany; Institute of Virology, University of Zurich, Zurich, Switzerland; Cardiology Division, Clinic of Internal Medicine I, Friedrich Schiller University, Jena, Germany; and European Institute for Molecular Imaging,
| | - Raphaela Richter
- From the Laboratory for Gene Therapy and Molecular Imaging at the Max Planck Institute for Neurological Research, Cologne, Germany; Department of Physiology, Faculty of Medicine, Justus Liebig University, Giessen, Germany; Institute for Neurophysiology, University of Cologne, Cologne, Germany; Institute of Virology, University of Zurich, Zurich, Switzerland; Cardiology Division, Clinic of Internal Medicine I, Friedrich Schiller University, Jena, Germany; and European Institute for Molecular Imaging,
| | - Heinrich Sauer
- From the Laboratory for Gene Therapy and Molecular Imaging at the Max Planck Institute for Neurological Research, Cologne, Germany; Department of Physiology, Faculty of Medicine, Justus Liebig University, Giessen, Germany; Institute for Neurophysiology, University of Cologne, Cologne, Germany; Institute of Virology, University of Zurich, Zurich, Switzerland; Cardiology Division, Clinic of Internal Medicine I, Friedrich Schiller University, Jena, Germany; and European Institute for Molecular Imaging,
| | - Jürgen Hescheler
- From the Laboratory for Gene Therapy and Molecular Imaging at the Max Planck Institute for Neurological Research, Cologne, Germany; Department of Physiology, Faculty of Medicine, Justus Liebig University, Giessen, Germany; Institute for Neurophysiology, University of Cologne, Cologne, Germany; Institute of Virology, University of Zurich, Zurich, Switzerland; Cardiology Division, Clinic of Internal Medicine I, Friedrich Schiller University, Jena, Germany; and European Institute for Molecular Imaging,
| | - Cornel Fraefel
- From the Laboratory for Gene Therapy and Molecular Imaging at the Max Planck Institute for Neurological Research, Cologne, Germany; Department of Physiology, Faculty of Medicine, Justus Liebig University, Giessen, Germany; Institute for Neurophysiology, University of Cologne, Cologne, Germany; Institute of Virology, University of Zurich, Zurich, Switzerland; Cardiology Division, Clinic of Internal Medicine I, Friedrich Schiller University, Jena, Germany; and European Institute for Molecular Imaging,
| | - Maria Wartenberg
- From the Laboratory for Gene Therapy and Molecular Imaging at the Max Planck Institute for Neurological Research, Cologne, Germany; Department of Physiology, Faculty of Medicine, Justus Liebig University, Giessen, Germany; Institute for Neurophysiology, University of Cologne, Cologne, Germany; Institute of Virology, University of Zurich, Zurich, Switzerland; Cardiology Division, Clinic of Internal Medicine I, Friedrich Schiller University, Jena, Germany; and European Institute for Molecular Imaging,
| | - Andreas H. Jacobs
- From the Laboratory for Gene Therapy and Molecular Imaging at the Max Planck Institute for Neurological Research, Cologne, Germany; Department of Physiology, Faculty of Medicine, Justus Liebig University, Giessen, Germany; Institute for Neurophysiology, University of Cologne, Cologne, Germany; Institute of Virology, University of Zurich, Zurich, Switzerland; Cardiology Division, Clinic of Internal Medicine I, Friedrich Schiller University, Jena, Germany; and European Institute for Molecular Imaging,
| |
Collapse
|
24
|
White E, Bienemann A, Megraw L, Bunnun C, Gill S. Evaluation and optimization of the administration of a selectively replicating herpes simplex viral vector to the brain by convection-enhanced delivery. Cancer Gene Ther 2011; 18:358-69. [PMID: 21372854 DOI: 10.1038/cgt.2011.2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The direct intraparenchymal administration of oncolytic viral vectors by convection-enhanced delivery (CED) represents a promising new treatment strategy for malignant gliomas. However, there is no evidence to suggest that oncolytic viruses as large as herpes simplex virus-1 (HSV-1) can be administered by CED, as this has not been systematically examined in an animal model. In this study, the administration of a herpes simplex viral vector, HSV1, has been evaluated in detail in the gray and white matter of both rat and pig models, using high flow-rate infusions, co-infusing heparin or preinfusing the tissue with an isotonic albumin solution. Rat HSV-1 infusions at both slow (0.5 μl min(-1)) and high infusion rates (2.5 μl min(-1)) led to extensive tissue damage and negligible cell transduction. Co-infusion with heparin led to extensive hemorrhage. Preinfusion of tissue with an isotonic albumin solution facilitated widespread vector distribution and cell transduction in white matter only. Using this approach in pig brain led to widespread vector distribution with extensive transduction of astrocytes and activated microglia. In rat brain, enhanced green fluorescent protein expression peaked 48 h after vector administration and was associated with a vigorous immune response. These findings indicate that direct infusions of HSV-1-based viral vectors into the brain lead to minimal vector distribution, negligible cell transduction and extensive damage. Tissue preinfusion with an isotonic solution prior to vector administration represents an effective technique for achieving widespread HSV-1 distribution.
Collapse
Affiliation(s)
- E White
- Department of Neurosurgery, Frenchay Hospital, Bristol, UK
| | | | | | | | | |
Collapse
|
25
|
Gaston DC, Whitley RJ, Parker JN. Engineered herpes simplex virus vectors for antitumor therapy and vaccine delivery. Future Virol 2011. [DOI: 10.2217/fvl.11.4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Genetically modified herpes simplex viruses (HSVs) have been exploited for both antitumor therapy and vaccine delivery. These mutant viruses retain their ability to replicate and lyse permissive cells, including many tumor types, and are referred to as oncolytic HSVs. In addition, deletion of nonessential genes permits the introduction of foreign genes to augment the antitumor effect by either immune stimulation, targeting for select tumors, or expression of tumor or vaccine antigens. This article reviews the development of oncolytic HSVs as an anticancer therapy, as well as the application of HSV-1 vectors for delivery of targeted antigens or as vaccine adjuvants. The impact of these novel vectors with respect to enhanced antitumor activity and development of antitumor vaccination strategies is discussed.
Collapse
Affiliation(s)
- David C Gaston
- Medical Scientist Training Program, Department of Cell Biology, CHB 130, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Richard J Whitley
- Departments of Pediatrics, Microbiology, Medicine & Neurosurgery, CHB 303, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Jacqueline N Parker
- Departments of Pediatrics & Cell Biology, CHB 118B, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| |
Collapse
|
26
|
Kanai R, Wakimoto H, Cheema T, Rabkin SD. Oncolytic herpes simplex virus vectors and chemotherapy: are combinatorial strategies more effective for cancer? Future Oncol 2010; 6:619-34. [PMID: 20373873 DOI: 10.2217/fon.10.18] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Despite aggressive treatments, including chemotherapy and radiotherapy, cancers often recur owing to resistance to conventional therapies. Oncolytic viruses such as oncolytic herpes simplex virus (oHSV) represent an exciting biological approach to cancer therapy. A range of viral mutations has been engineered into HSV to engender oncolytic activity. While oHSV as a single agent has been tested in a number of cancer clinical trials, preclinical studies have demonstrated enhanced efficacy when it is combined with cytotoxic anticancer drugs. Among the strategies that will be discussed in this article are combinations with standard-of-care chemotherapeutics, expression of prodrug-activating enzymes to enhance chemotherapy and small-molecule inhibitors. The combination of oHSV and chemotherapy can achieve much more efficient cancer cell killing than either single agent alone, often through synergistic interactions. This can be clinically important not just for improving efficacy but also for permitting lower and less toxic chemotherapeutic doses. The viral mutations in an oHSV vector often determine the favorability of its interactions with chemotherapy, just as different cancer cells, due to genetic alterations, vary in their response to chemotherapy. As chemotherapeutics are often the standard of care, combining them with an investigational new drug, such as oHSV, is clinically easier than combining multiple novel agents. As has become clear for most cancer therapies, multimodal treatments are usually more effective. In this article, we will discuss the recent progress of these combinatorial strategies between virotherapy and chemotherapy and future directions.
Collapse
Affiliation(s)
- Ryuichi Kanai
- Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital, & Harvard Medical School, Boston, MA, USA
| | | | | | | |
Collapse
|
27
|
Hadjipanayis CG, Machaidze R, Kaluzova M, Wang L, Schuette AJ, Chen H, Wu X, Mao H. EGFRvIII antibody-conjugated iron oxide nanoparticles for magnetic resonance imaging-guided convection-enhanced delivery and targeted therapy of glioblastoma. Cancer Res 2010; 70:6303-12. [PMID: 20647323 DOI: 10.1158/0008-5472.can-10-1022] [Citation(s) in RCA: 272] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The magnetic nanoparticle has emerged as a potential multifunctional clinical tool that can provide cancer cell detection by magnetic resonance imaging (MRI) contrast enhancement as well as targeted cancer cell therapy. A major barrier in the use of nanotechnology for brain tumor applications is the difficulty in delivering nanoparticles to intracranial tumors. Iron oxide nanoparticles (IONP; 10 nm in core size) conjugated to a purified antibody that selectively binds to the epidermal growth factor receptor (EGFR) deletion mutant (EGFRvIII) present on human glioblastoma multiforme (GBM) cells were used for therapeutic targeting and MRI contrast enhancement of experimental glioblastoma, both in vitro and in vivo, after convection-enhanced delivery (CED). A significant decrease in glioblastoma cell survival was observed after nanoparticle treatment and no toxicity was observed with treatment of human astrocytes (P < 0.001). Lower EGFR phosphorylation was found in glioblastoma cells after EGFRvIIIAb-IONP treatment. Apoptosis was determined to be the mode of cell death after treatment of GBM cells and glioblastoma stem cell-containing neurospheres with EGFRvIIIAb-IONPs. MRI-guided CED of EGFRvIIIAb-IONPs allowed for the initial distribution of magnetic nanoparticles within or adjacent to intracranial human xenograft tumors and continued dispersion days later. A significant increase in animal survival was found after CED of magnetic nanoparticles (P < 0.01) in mice implanted with highly tumorigenic glioblastoma xenografts (U87DeltaEGFRvIII). IONPs conjugated to an antibody specific to the EGFRvIII deletion mutant constitutively expressed by human glioblastoma tumors can provide selective MRI contrast enhancement of tumor cells and targeted therapy of infiltrative glioblastoma cells after CED.
Collapse
Affiliation(s)
- Costas G Hadjipanayis
- Brain Tumor Nanotechnology Laboratory, Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia 30322, USA.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Cassady KA, Parker JN. Herpesvirus vectors for therapy of brain tumors. Open Virol J 2010; 4:103-8. [PMID: 20811578 DOI: 10.2174/1874357901004030103] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Revised: 01/07/2010] [Accepted: 01/07/2010] [Indexed: 11/22/2022] Open
Abstract
Genetically modified, conditionally-replicating Herpes Simplex Virus Type 1 (HSV-1) vectors for the treatment of malignant glioma have provided encouraging results in the handful of Phase I and Phase II clinical trials conducted to date. In recent years, a number of new strategies have been developed to improve anti-tumor activity of these attenuated vectors, through either introduction of foreign gene inserts to enhance tumor killing through a variety of mechanisms, or through combination with existing treatment regimens, including radiation and/or chemotherapeutics. Another promising new approach has been the engineering of novel oncolytic HSV vectors that retain wildtype replication, but are targeted to tumor cells through a variety of mechanisms. This review summarizes the latest advances in herpesvirus-mediated oncolytic therapies from both preclinical results and clinical trials with oncolytic HSV vectors in patients, and their implication for design of future trials.
Collapse
Affiliation(s)
- Kevin A Cassady
- Department of Pediatrics, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, AL 35294-0011, USA
| | | |
Collapse
|
29
|
Luther N, Cheung NK, Souliopoulos EP, Karampelas I, Karempelas I, Bassiri D, Edgar MA, Guo HF, Pastan I, Gutin PH, Souweidane MM. Interstitial infusion of glioma-targeted recombinant immunotoxin 8H9scFv-PE38. Mol Cancer Ther 2010; 9:1039-46. [PMID: 20371725 DOI: 10.1158/1535-7163.mct-09-0996] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Monoclonal antibodies have the potential to target therapy for high-grade gliomas. Monoclonal antibody 8H9 is specific for membrane protein B7H3 and is reactive with most human high-grade gliomas. We tested the 8H9scFv-PE38 recombinant Pseudomonas immunotoxin in a preclinical model of high-grade glioma. The half maximal inhibitory concentration (IC(50)) of 8H9scFv-PE38 in vitro was determined using glioblastoma cell lines U87 and U251. Maximum tolerated infusion dose of 8H9scFv-PE38 following interstitial infusion to the striatum and pons was defined using athymic rats. Maximum tolerated infusion dose of 8H9scFv-PE38 or PBS control were interstitially delivered to athymic rats xenografted with U87 in the striatum or brain stem. Radiographic response and survivals were measured and compared between treatment groups. The in vitro IC(50) of 8H9scFv-PE38 for U87 was 1,265 ng/mL and, for U251, 91 ng/mL. The maximum tolerated infusion doses of interstitially infused 8H9scFv-PE38 to the striatum and brain stem were 0.75 and 1.8 mug, respectively. For rats harboring intracranial U87 xenografts, infusion of 8H9scFv-PE38 increased mean survival (striatum, 43.4 versus 24.6 days; brain stem, 80.6 versus 45.5 days; n = 28 total) and produced three long-term survivors past 120 days. None of the 14 placebo-treated animals survived >54 days. Tumors also showed volumetric response to infusion of 8H9scFv-PE38 by magnetic resonance imaging. Interstitial infusion of 8H9scFv-PE38 shows potential for the treatment of hemispherical and brain stem glioma. Mol Cancer Ther; 9(4); 1039-46. (c)2010 AACR.
Collapse
Affiliation(s)
- Neal Luther
- Department of Neurologic Surgery, Weill Medical College of Cornell University, Room A-969, 1300 York Avenue, New York, NY, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Debinski W, Tatter SB. Convection-enhanced delivery for the treatment of brain tumors. Expert Rev Neurother 2009; 9:1519-27. [PMID: 19831841 DOI: 10.1586/ern.09.99] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The brain is highly accessible for nutrients and oxygen, however delivery of drugs to malignant brain tumors is a very challenging task. Convection-enhanced delivery (CED) has been designed to overcome some of the difficulties so that pharmacological agents that would not normally cross the BBB can be used for treatment. Drugs are delivered through one to several catheters placed stereotactically directly within the tumor mass or around the tumor or the resection cavity. Several classes of drugs are amenable to this technology including standard chemotherapeutics or novel experimental targeted drugs. The first Phase III trial for CED-delivered, molecularly targeted cytotoxin in the treatment of recurrent glioblastoma multiforme has been accomplished and demonstrated objective clinical efficacy. The lessons learned from more than a decade of attempts at exploiting CED for brain cancer treatment weigh critically for its future clinical applications. The main issues center around the type of catheters used, number of catheters and their exact placement; pharmacological formulation of drugs, prescreening patients undergoing treatment and monitoring the distribution of drugs in tumors and the tumor-infiltrated brain. It is expected that optimizing CED will make this technology a permanent addition to clinical management of brain malignancies.
Collapse
Affiliation(s)
- Waldemar Debinski
- Brain Tumor Center of Excellence, Comprehensive Cancer Center of Wake Forest University, Department of Neurosurgery, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA.
| | | |
Collapse
|
31
|
Parker JN, Bauer DF, Cody JJ, Markert JM. Oncolytic viral therapy of malignant glioma. Neurotherapeutics 2009; 6:558-69. [PMID: 19560745 PMCID: PMC3980727 DOI: 10.1016/j.nurt.2009.04.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2009] [Accepted: 04/24/2009] [Indexed: 10/20/2022] Open
Abstract
Novel approaches to treatment of malignant glioma, the most frequently occurring primary brain tumor, have included the use of a wide range of oncolytic viral vectors. These vectors, either naturally tumor-selective, or engineered as such, have shown promise in the handful of phase I and phase II clinical trials conducted in recent years. The strategies developed for each of the different viruses currently being studied and the history of their development are summarized here. In addition, the results of clinical trials in patients and their implication for future trials are also discussed.
Collapse
Affiliation(s)
- Jacqueline Nuss Parker
- grid.265892.20000000106344187Department of Pediatrics, Division of Infectious Diseases, University of Alabama at Birmingham, 35294 Birmingham, Alabama
| | - David F. Bauer
- grid.265892.20000000106344187Department of Surgery, Division of Neurosurgery, University of Alabama at Birmingham, 35294 Birmingham, Alabama
| | - James J. Cody
- grid.265892.20000000106344187Department of Pediatrics, Division of Infectious Diseases, University of Alabama at Birmingham, 35294 Birmingham, Alabama
| | - James M. Markert
- grid.265892.20000000106344187Department of Surgery, Division of Neurosurgery, University of Alabama at Birmingham, 35294 Birmingham, Alabama
| |
Collapse
|